1
|
He Q, Wang Z, Wang Y, Zhu M, Liang Z, Zhang K, Xu Y, Chen G. Characteristic changes in astrocyte properties during astrocyte-to-neuron conversion induced by NeuroD1/Ascl1/Dlx2. Neural Regen Res 2025; 20:1801-1815. [PMID: 39104117 PMCID: PMC11688565 DOI: 10.4103/nrr.nrr-d-23-01897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/04/2024] [Accepted: 03/25/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00030/figure1/v/2024-08-05T133530Z/r/image-tiff Direct in vivo conversion of astrocytes into functional new neurons induced by neural transcription factors has been recognized as a potential new therapeutic intervention for neural injury and degenerative disorders. However, a few recent studies have claimed that neural transcription factors cannot convert astrocytes into neurons, attributing the converted neurons to pre-existing neurons mis-expressing transgenes. In this study, we overexpressed three distinct neural transcription factors--NeuroD1, Ascl1, and Dlx2--in reactive astrocytes in mouse cortices subjected to stab injury, resulting in a series of significant changes in astrocyte properties. Initially, the three neural transcription factors were exclusively expressed in the nuclei of astrocytes. Over time, however, these astrocytes gradually adopted neuronal morphology, and the neural transcription factors was gradually observed in the nuclei of neuron-like cells instead of astrocytes. Furthermore, we noted that transcription factor-infected astrocytes showed a progressive decrease in the expression of astrocytic markers AQP4 (astrocyte endfeet signal), CX43 (gap junction signal), and S100β. Importantly, none of these changes could be attributed to transgene leakage into pre-existing neurons. Therefore, our findings suggest that neural transcription factors such as NeuroD1, Ascl1, and Dlx2 can effectively convert reactive astrocytes into neurons in the adult mammalian brain.
Collapse
Affiliation(s)
- Qing He
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhen Wang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Yuchen Wang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Mengjie Zhu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhile Liang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Kanghong Zhang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Yuge Xu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Gong Chen
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Snapyan M, Desmeules F, Munro J, Bérard M, Saikali S, Gould PV, Richer M, Pourcher E, Langlois M, Dufresne A, Prud'homme M, Cantin L, Parent A, Saghatelyan A, Parent M. Adult Neurogenesis in the Subventricular Zone of Patients with Huntington's and Parkinson's Diseases and following Long-Term Treatment with Deep Brain Stimulation. Ann Neurol 2025; 97:894-906. [PMID: 39829080 PMCID: PMC12010058 DOI: 10.1002/ana.27181] [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: 07/24/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Parkinson's and Huntington's diseases are characterized by progressive neuronal loss. Previous studies using human postmortem tissues have shown the impact of neurodegenerative disorders on adult neurogenesis. The extent to which adult neural stem cells are activated in the subventricular zone and whether therapeutic treatments such as deep brain stimulation promote adult neurogenesis remains unclear. The goal of the present study is to assess adult neural stem cells activation and neurogenesis in the subventricular zone of patients with Huntington's and Parkinson's diseases who were treated or not by deep brain stimulation. METHODS Postmortem brain samples from Huntington's and Parkinson's disease patients who had received or not long-term deep brain stimulation of the subthalamic nucleus were used. RESULTS Our results indicate a significant increase in the thickness of the subventricular zone and in the density of proliferating cells and activated stem cells in the brain of Huntington's disease subjects and Parkinson's disease patients treated with deep brain stimulation. We also observed an increase in the density of immature neurons in the brain of these patients. INTERPRETATION Overall, our data indicate that long-term deep brain stimulation of the subthalamic nucleus promotes cell proliferation and neurogenesis in the subventricular zone that are reduced in Parkinson's disease. Taken together, our results also provide a detailed characterization of the cellular composition of the adult human subventricular zone and caudate nucleus in normal condition and in Parkinson's and Huntington's diseases and demonstrate the plasticity of these regions in response to neurodegeneration. ANN NEUROL 2025;97:894-906.
Collapse
Affiliation(s)
| | - Francis Desmeules
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | | | | | - Stephan Saikali
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Peter V. Gould
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Maxime Richer
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Emmanuelle Pourcher
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Mélanie Langlois
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Anne‐Marie Dufresne
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Michel Prud'homme
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Léo Cantin
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - André Parent
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Department of Psychiatry and NeuroscienceFaculty of Medicine, Université LavalQuebec CityQuebecCanada
| | - Armen Saghatelyan
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Department of Psychiatry and NeuroscienceFaculty of Medicine, Université LavalQuebec CityQuebecCanada
- Department of Cellular and Molecular MedicineFaculty of Medicine, University of OttawaOttawaOntarioCanada
| | - Martin Parent
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Department of Psychiatry and NeuroscienceFaculty of Medicine, Université LavalQuebec CityQuebecCanada
| |
Collapse
|
3
|
Feng Z, Zuo Y, Shen J, Zhao Q, Cao ZQ, Li X, Wang Z. Bioengineering microspheres regulating mesenchymal stem cell fate accelerate spinal cord injury therapeutics. NANO TODAY 2025; 61:102574. [DOI: 10.1016/j.nantod.2024.102574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Zeng CW. Immune Cell-NSPC interactions: Friend or foe in CNS injury and repair? Differentiation 2025; 143:100855. [PMID: 40112742 DOI: 10.1016/j.diff.2025.100855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Neural stem/progenitor cells (NSPCs) play a crucial role in central nervous system (CNS) development, regeneration, and repair. However, their functionality and therapeutic potential are intricately modulated by interactions with immune cells, particularly macrophages and microglia. Microglia, as CNS-resident macrophages, are distinct from peripheral macrophages in their roles and characteristics, contributing to specialized functions within the CNS. Recent evidence suggests that microglia, as CNS-resident macrophages, contribute to the quality assurance of NSPCs by eliminating stressed or dysfunctional cells, yet the mechanisms underlying this process remain largely unexplored. Furthermore, macrophage polarization states, such as M1 and M2, appear to differentially influence NSPC quality, potentially impacting neurogenesis and regenerative outcomes. Identifying surface markers indicative of NSPC stress could provide a strategy for selecting optimal cells for transplantation therapies. Additionally, in vivo clonal labeling approaches may enable precise tracking of NSPC fate and their interactions with immune cells. Beyond macrophages and microglia, the roles of other immune cells, including T cells and neutrophils, particularly in injury and neurodegenerative disease contexts, in the context of CNS injury and disease are emerging areas of interest. Here, I discuss the emerging evidence supporting the interplay between the immune system and NSPCs, highlighting critical gaps in knowledge and proposing future research directions to harness immune-mediated mechanisms for optimizing neural regeneration and transplantation strategies.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
5
|
Li T, Xing HM, Qian HD, Gao Q, Xu SL, Ma H, Chi ZL. Small extracellular vesicles derived from human induced pluripotent stem cell-differentiated neural progenitor cells mitigate retinal ganglion cell degeneration in a mouse model of optic nerve injury. Neural Regen Res 2025; 20:587-597. [PMID: 38819069 PMCID: PMC11317950 DOI: 10.4103/nrr.nrr-d-23-01414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00034/figure1/v/2024-05-28T214302Z/r/image-tiff Several studies have found that transplantation of neural progenitor cells (NPCs) promotes the survival of injured neurons. However, a poor integration rate and high risk of tumorigenicity after cell transplantation limits their clinical application. Small extracellular vesicles (sEVs) contain bioactive molecules for neuronal protection and regeneration. Previous studies have shown that stem/progenitor cell-derived sEVs can promote neuronal survival and recovery of neurological function in neurodegenerative eye diseases and other eye diseases. In this study, we intravitreally transplanted sEVs derived from human induced pluripotent stem cells (hiPSCs) and hiPSCs-differentiated NPCs (hiPSC-NPC) in a mouse model of optic nerve crush. Our results show that these intravitreally injected sEVs were ingested by retinal cells, especially those localized in the ganglion cell layer. Treatment with hiPSC-NPC-derived sEVs mitigated optic nerve crush-induced retinal ganglion cell degeneration, and regulated the retinal microenvironment by inhibiting excessive activation of microglia. Component analysis further revealed that hiPSC-NPC derived sEVs transported neuroprotective and anti-inflammatory miRNA cargos to target cells, which had protective effects on RGCs after optic nerve injury. These findings suggest that sEVs derived from hiPSC-NPC are a promising cell-free therapeutic strategy for optic neuropathy.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hui-Min Xing
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hai-Dong Qian
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qiao Gao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Sheng-Lan Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hua Ma
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zai-Long Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
6
|
Luciani M, Garsia C, Beretta S, Cifola I, Peano C, Merelli I, Petiti L, Miccio A, Meneghini V, Gritti A. Human iPSC-derived neural stem cells displaying radial glia signature exhibit long-term safety in mice. Nat Commun 2024; 15:9433. [PMID: 39487141 PMCID: PMC11530573 DOI: 10.1038/s41467-024-53613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NSCs) hold promise for treating neurodegenerative and demyelinating disorders. However, comprehensive studies on their identity and safety remain limited. In this study, we demonstrate that hiPSC-NSCs adopt a radial glia-associated signature, sharing key epigenetic and transcriptional characteristics with human fetal neural stem cells (hfNSCs) while exhibiting divergent profiles from glioblastoma stem cells. Long-term transplantation studies in mice showed robust and stable engraftment of hiPSC-NSCs, with predominant differentiation into glial cells and no evidence of tumor formation. Additionally, we identified the Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) as a regulator of astroglial differentiation in hiPSC-NSCs. These findings provide valuable transcriptional and epigenetic reference datasets to prospectively define the maturation stage of NSCs derived from different hiPSC sources and demonstrate the long-term safety of hiPSC-NSCs, reinforcing their potential as a viable alternative to hfNSCs for clinical applications.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Garsia
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Beretta
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Rozzano, Milan, Italy
- Human Technopole, Via Rita Levi Montalcini 1, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Luca Petiti
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Annarita Miccio
- IMAGINE Institute, Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
7
|
Xue J, Chu Y, Huang Y, Chen M, Sun M, Fan Z, Wu Y, Chen L. A tumorigenicity evaluation platform for cell therapies based on brain organoids. Transl Neurodegener 2024; 13:53. [PMID: 39472972 PMCID: PMC11520457 DOI: 10.1186/s40035-024-00446-5] [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: 05/27/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Tumorigenicity represents a critical challenge in stem cell-based therapies requiring rigorous monitoring. Conventional approaches for tumorigenicity evaluation are based on animal models and have numerous limitations. Brain organoids, which recapitulate the structural and functional complexity of the human brain, have been widely used in neuroscience research. However, the capacity of brain organoids for tumorigenicity evaluation needs to be further elucidated. METHODS A cerebral organoid model produced from human pluripotent stem cells (hPSCs) was employed. Meanwhile, to enhance the detection sensitivity for potential tumorigenic cells, we created a glioblastoma-like organoid (GBM organoid) model from TP53-/-/PTEN-/- hPSCs to provide a tumor microenvironment for injected cells. Midbrain dopamine (mDA) cells from human embryonic stem cells were utilized as a cell therapy product. mDA cells, hPSCs, mDA cells spiked with hPSCs, and immature mDA cells were then injected into the brain organoids and NOD SCID mice. The injected cells within the brain organoids were characterized, and compared with those injected in vivo to evaluate the capability of the brain organoids for tumorigenicity evaluation. Single-cell RNA sequencing was performed to identify the differential gene expression between the cerebral organoids and the GBM organoids. RESULTS Both cerebral organoids and GBM organoids supported maturation of the injected mDA cells. The hPSCs and immature mDA cells injected in the GBM organoids showed a significantly higher proliferative capacity than those injected in the cerebral organoids and in NOD SCID mice. Furthermore, the spiked hPSCs were detectable in both the cerebral organoids and the GBM organoids. Notably, the GBM organoids demonstrated a superior capacity to enhance proliferation and pluripotency of spiked hPSCs compared to the cerebral organoids and the mouse model. Kyoto Encyclopedia of Genes and Genomes analysis revealed upregulation of tumor-related metabolic pathways and cytokines in the GBM organoids, suggesting that these factors underlie the high detection sensitivity for tumorigenicity evaluation. CONCLUSIONS Our findings suggest that brain organoids could represent a novel and effective platform for evaluating the tumorigenic risk in stem cell-based therapies. Notably, the GBM organoids offer a superior platform that could complement or potentially replace traditional animal-based models for tumorigenicity evaluation.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yanwang Huang
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ming Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Zhiqin Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
8
|
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
|
9
|
Yuan M, Tang Y, Huang T, Ke L, Huang E. In situ direct reprogramming of astrocytes to neurons via polypyrimidine tract-binding protein 1 knockdown in a mouse model of ischemic stroke. Neural Regen Res 2024; 19:2240-2248. [PMID: 38488558 PMCID: PMC11034579 DOI: 10.4103/1673-5374.390957] [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: 05/06/2023] [Revised: 08/09/2023] [Accepted: 10/16/2023] [Indexed: 04/24/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202410000-00025/figure1/v/2024-02-06T055622Z/r/image-tiff In situ direct reprogramming technology can directly convert endogenous glial cells into functional neurons in vivo for central nervous system repair. Polypyrimidine tract-binding protein 1 (PTB) knockdown has been shown to reprogram astrocytes to functional neurons in situ. In this study, we used AAV-PHP.eB-GFAP-shPTB to knockdown PTB in a mouse model of ischemic stroke induced by endothelin-1, and investigated the effects of GFAP-shPTB-mediated direct reprogramming to neurons. Our results showed that in the mouse model of ischemic stroke, PTB knockdown effectively reprogrammed GFAP-positive cells to neurons in ischemic foci, restored neural tissue structure, reduced inflammatory response, and improved behavioral function. These findings validate the effectiveness of in situ transdifferentiation of astrocytes, and suggest that the approach may be a promising strategy for stroke treatment.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yao Tang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Tianwen Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lining Ke
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - En Huang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
10
|
Eivazi Zadeh Z, Nour S, Kianersi S, Jonidi Shariatzadeh F, Williams RJ, Nisbet DR, Bruggeman KF. Mining human clinical waste as a rich source of stem cells for neural regeneration. iScience 2024; 27:110307. [PMID: 39156636 PMCID: PMC11326931 DOI: 10.1016/j.isci.2024.110307] [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] [Indexed: 08/20/2024] Open
Abstract
Neural diseases are challenging to treat and are regarded as one of the major causes of disability and morbidity in the world. Stem cells can provide a solution, by offering a mechanism to replace damaged circuitry. However, obtaining sufficient cell sources for neural regeneration remains a significant challenge. In recent years, waste-derived stem(-like) cells (WDS-lCs) extracted from both prenatal and adult clinical waste tissues/products, have gained increasing attention for application in neural tissue repair and remodeling. This often-overlooked pool of cells possesses favorable characteristics; including self-renewal, neural differentiation, secretion of neurogenic factors, cost-effectiveness, and low ethical concerns. Here, we offer a perspective regarding the biological properties, extraction protocols, and preclinical and clinical treatments where prenatal and adult WDS-lCs have been utilized for cell replacement therapy in neural applications, and the challenges involved in optimizing these approaches toward patient led therapies.
Collapse
Affiliation(s)
- Zahra Eivazi Zadeh
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Shirin Nour
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- Polymer Science Group, Department of Chemical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sogol Kianersi
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences, University of Galway, Galway, Ireland
| | | | - Richard J. Williams
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- iMPACT, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - David R. Nisbet
- Department of Biomedical Engineering, University of Melbourne, Parkville, VIC 3010, Australia
- The Graeme Clark Institute, University of Melbourne, Melbourne, VIC, Australia
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU College of Health & Medicine, Canberra, ACT, Australia
- Research School of Chemistry, ANU College of Science, Canberra, ACT, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC, Australia
- Founder and Scientific Advisory of Nano Status, Building 137, Sullivans Creek Rd, ANU, Acton, Canberra, ACT, Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research, School of Engineering, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
11
|
Xiang Z, He S, Chen R, Liu S, Liu M, Xu L, Zheng J, Jiang Z, Ma L, Sun Y, Qin Y, Chen Y, Li W, Wang X, Chen G, Lei W. Two-photon live imaging of direct glia-to-neuron conversion in the mouse cortex. Neural Regen Res 2024; 19:1781-1788. [PMID: 38103245 PMCID: PMC10960291 DOI: 10.4103/1673-5374.386401] [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: 04/28/2023] [Revised: 08/23/2023] [Accepted: 09/26/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00032/figure1/v/2023-12-16T180322Z/r/image-tiff Over the past decade, a growing number of studies have reported transcription factor-based in situ reprogramming that can directly convert endogenous glial cells into functional neurons as an alternative approach for neuroregeneration in the adult mammalian central nervous system. However, many questions remain regarding how a terminally differentiated glial cell can transform into a delicate neuron that forms part of the intricate brain circuitry. In addition, concerns have recently been raised around the absence of astrocyte-to-neuron conversion in astrocytic lineage-tracing mice. In this study, we employed repetitive two-photon imaging to continuously capture the in situ astrocyte-to-neuron conversion process following ectopic expression of the neural transcription factor NeuroD1 in both proliferating reactive astrocytes and lineage-traced astrocytes in the mouse cortex. Time-lapse imaging over several weeks revealed the step-by-step transition from a typical astrocyte with numerous short, tapered branches to a typical neuron with a few long neurites and dynamic growth cones that actively explored the local environment. In addition, these lineage-converting cells were able to migrate radially or tangentially to relocate to suitable positions. Furthermore, two-photon Ca2+ imaging and patch-clamp recordings confirmed that the newly generated neurons exhibited synchronous calcium signals, repetitive action potentials, and spontaneous synaptic responses, suggesting that they had made functional synaptic connections within local neural circuits. In conclusion, we directly visualized the step-by-step lineage conversion process from astrocytes to functional neurons in vivo and unambiguously demonstrated that adult mammalian brains are highly plastic with respect to their potential for neuroregeneration and neural circuit reconstruction.
Collapse
Affiliation(s)
- Zongqin Xiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- Laboratory for Neuroimmunology in Health and Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Shu He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Rongjie Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Shanggong Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Minhui Liu
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Flemish Region, Belgium
| | - Liang Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Jiajun Zheng
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zhouquan Jiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Long Ma
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Ying Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yongpeng Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Yi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wen Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
12
|
Kumar Podder A, Mohamed MA, Seidman RA, Tseropoulos G, Polanco JJ, Lei P, Sim FJ, Andreadis ST. Injectable shear-thinning hydrogels promote oligodendrocyte progenitor cell survival and remyelination in the central nervous system. SCIENCE ADVANCES 2024; 10:eadk9918. [PMID: 38996029 PMCID: PMC11244542 DOI: 10.1126/sciadv.adk9918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 06/10/2024] [Indexed: 07/14/2024]
Abstract
Cell therapy for the treatment of demyelinating diseases such as multiple sclerosis is hampered by poor survival of donor oligodendrocyte cell preparations, resulting in limited therapeutic outcomes. Excessive cell death leads to the release of intracellular alloantigens, which likely exacerbate local inflammation and may predispose the graft to eventual rejection. Here, we engineered innovative cell-instructive shear-thinning hydrogels (STHs) with tunable viscoelasticity and bioactivity for minimally invasive delivery of primary human oligodendrocyte progenitor cells (hOPCs) to the brain of a shiverer/rag2 mouse, a model of congenital hypomyelinating disease. The STHs enabled immobilization of prosurvival signals, including a recombinantly designed bidomain peptide and platelet-derived growth factor. Notably, STHs reduced the death rate of hOPCs significantly, promoted the production of myelinating oligodendrocytes, and enhanced myelination of the mouse brain 12 weeks post-implantation. Our results demonstrate the potential of STHs loaded with biological cues to improve cell therapies for the treatment of devastating myelopathies.
Collapse
Affiliation(s)
- Ashis Kumar Podder
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
| | - Mohamed Alaa Mohamed
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
| | - Richard A. Seidman
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
| | - Jessie J. Polanco
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Pedro Lei
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
| | - Fraser J. Sim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, USA
- Department of Biomedical Engineering, University at Buffalo, SUNY, Buffalo, NY, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
- Center of Cell, Gene and Tissue Engineering, University at Buffalo, SUNY, Buffalo, NY, USA
| |
Collapse
|
13
|
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
|
14
|
Alzahrani FA, Riza YM, Eid TM, Almotairi R, Scherschinski L, Contreras J, Nadeem M, Perez SE, Raikwar SP, Jha RM, Preul MC, Ducruet AF, Lawton MT, Bhatia K, Akhter N, Ahmad S. Exosomes in Vascular/Neurological Disorders and the Road Ahead. Cells 2024; 13:670. [PMID: 38667285 PMCID: PMC11049650 DOI: 10.3390/cells13080670] [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: 03/22/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), stroke, and aneurysms, are characterized by the abnormal accumulation and aggregation of disease-causing proteins in the brain and spinal cord. Recent research suggests that proteins linked to these conditions can be secreted and transferred among cells using exosomes. The transmission of abnormal protein buildup and the gradual degeneration in the brains of impacted individuals might be supported by these exosomes. Furthermore, it has been reported that neuroprotective functions can also be attributed to exosomes in neurodegenerative diseases. The potential neuroprotective functions may play a role in preventing the formation of aggregates and abnormal accumulation of proteins associated with the disease. The present review summarizes the roles of exosomes in neurodegenerative diseases as well as elucidating their therapeutic potential in AD, PD, ALS, HD, stroke, and aneurysms. By elucidating these two aspects of exosomes, valuable insights into potential therapeutic targets for treating neurodegenerative diseases may be provided.
Collapse
Affiliation(s)
- Faisal A. Alzahrani
- Department of Biochemistry, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Yasir M. Riza
- Department of Biochemistry, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Thamir M. Eid
- Department of Biochemistry, King Fahad Center for Medical Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Reema Almotairi
- Department of Medical Laboratory Technology, Prince Fahad bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Lea Scherschinski
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Jessica Contreras
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Muhammed Nadeem
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Sylvia E. Perez
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Sudhanshu P. Raikwar
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
| | - Ruchira M. Jha
- Department of Neurology, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Mark C. Preul
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Andrew F. Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Michael T. Lawton
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Kanchan Bhatia
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| | - Naseem Akhter
- Department of Biology, Arizona State University, Lake Havasu City, AZ 86403, USA
| | - Saif Ahmad
- Department of Translational Neuroscience, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA (J.C.)
- Department of Neurosurgery, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
- Phoenix Veterans Affairs (VA) Health Care System, Phoenix, AZ 85012, USA
| |
Collapse
|
15
|
Adewumi HO, Berniac GI, McCarthy EA, O'Shea TM. Ischemic and hemorrhagic stroke lesion environments differentially alter the glia repair potential of neural progenitor cell and immature astrocyte grafts. Exp Neurol 2024; 374:114692. [PMID: 38244885 DOI: 10.1016/j.expneurol.2024.114692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Using cell grafting to direct glia-based repair mechanisms in adult CNS injuries represents a potential therapeutic strategy for supporting functional neural parenchymal repair. However, glia repair directed by neural progenitor cell (NPC) grafts is dramatically altered by increasing lesion size, severity, and mode of injury. To address this, we studied the interplay between astrocyte differentiation and cell proliferation of NPC in vitro to generate proliferating immature astrocytes (ImA) using hysteretic conditioning. ImA maintain proliferation rates at comparable levels to NPC but showed robust immature astrocyte marker expression including Gfap and Vimentin. ImA demonstrated enhanced resistance to myofibroblast-like phenotypic transformations upon exposure to serum enriched environments in vitro compared to NPC and were more effective at scratch wound closure in vitro compared to quiescent astrocytes. Glia repair directed by ImA at acute ischemic striatal stroke lesions was equivalent to NPC but better than quiescent astrocyte grafts. While ischemic injury environments supported enhanced survival of grafts compared to healthy striatum, hemorrhagic lesions were hostile towards both NPC and ImA grafts leading to poor survival and ineffective modulation of natural wound repair processes. Our findings demonstrate that lesion environments, rather than transcriptional pre-graft states, determine the survival, cell-fate, and glia repair competency of cell grafts applied to acute CNS injuries.
Collapse
Affiliation(s)
- Honour O Adewumi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA
| | - Gabriela I Berniac
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA
| | - Emily A McCarthy
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA
| | - Timothy M O'Shea
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA.
| |
Collapse
|
16
|
Rogujski P, Lukomska B, Janowski M, Stanaszek L. Glial-restricted progenitor cells: a cure for diseased brain? Biol Res 2024; 57:8. [PMID: 38475854 DOI: 10.1186/s40659-024-00486-1] [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: 03/03/2023] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The central nervous system (CNS) is home to neuronal and glial cells. Traditionally, glia was disregarded as just the structural support across the brain and spinal cord, in striking contrast to neurons, always considered critical players in CNS functioning. In modern times this outdated dogma is continuously repelled by new evidence unravelling the importance of glia in neuronal maintenance and function. Therefore, glia replacement has been considered a potentially powerful therapeutic strategy. Glial progenitors are at the center of this hope, as they are the source of new glial cells. Indeed, sophisticated experimental therapies and exciting clinical trials shed light on the utility of exogenous glia in disease treatment. Therefore, this review article will elaborate on glial-restricted progenitor cells (GRPs), their origin and characteristics, available sources, and adaptation to current therapeutic approaches aimed at various CNS diseases, with particular attention paid to myelin-related disorders with a focus on recent progress and emerging concepts. The landscape of GRP clinical applications is also comprehensively presented, and future perspectives on promising, GRP-based therapeutic strategies for brain and spinal cord diseases are described in detail.
Collapse
Affiliation(s)
- Piotr Rogujski
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, USA
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| |
Collapse
|
17
|
Min XL, Liu HJ, Dou XK, Chen FX, Zhao Q, Zhao XH, Shi Y, Zhao QY, Sun SJ, Wang Z, Yu SH. Extracellular Vesicles from Neural Stem Cells Carry microRNA-16-5p to Reduce Corticosterone-induced Neuronal Injury in Depression Rats. Neuroscience 2024; 538:95-109. [PMID: 37778691 DOI: 10.1016/j.neuroscience.2023.09.016] [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: 02/28/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE Depression is a common mental illness. Neural stem cell-derived extracellular vesicles (NSC-EVs) are involved in repairing neuronal injury. We estimated the mechanism of miR-16-5p in depression rats. METHODS EVs were extracted from NSCs. The depression rat model was established by corticosterone (CORT) induction and treated with NSC-EVs. The depression behavioral/pathological changes in rats were assessed using forced swimming test, open field test, sucrose consumption test and western blotting. The neuronal apoptosis in hippocampal tissue were detected. CORT-induced PC12 cell model was established. EV uptake by PC12 cells was measured and PC12 cell apoptosis was detected. The downstream targets of miR-16-5p were predicted and verified. The expressions of miR-16-5p and MYB in rats, PC12 cells, and EVs were measured. Functional rescue experiments were conducted to verify the role of miR-16-5p and MYB in PC12 cell apoptosis. RESULTS CORT induction increased neuronal apoptosis in hippocampal tissue and induced depression-like behaviors in rats, while NSC-EV treatment improved depression-like behaviors and apoptosis in rats. In PC12 cells, NSC-EVs decreased CORT-induced PC12 cell apoptosis. NSC-EVs carried miR-16-5p into PC12 cells. miR-16-5p knockdown in EVs partially reversed the inhibitory effects of NSC-EVs on CORT-induced PC12 cell apoptosis. miR-16-5p targeted to inhibit MYB to repress CORT-induced PC12 cell apoptosis. In vivo experiments further verified that NSC-EVs reduced neuronal injury in CORT-induced depression rats via the miR-16-5p/MYB axis. CONCLUSION NSC-EVs-mediated alleviation on neuronal injury by carrying miR-16-5p to target MYB was highly likely one of the mechanisms by which NSC-EVs mediated miR-16-5p in neuroprotection of depression rats.
Collapse
Affiliation(s)
- Xiao-Li Min
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Hai-Jing Liu
- Department of Acupuncture and Massage, Yunnan Traditional Chinese Medicine University, Kunming, China
| | - Xing-Kui Dou
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei-Xiong Chen
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qing Zhao
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Hong Zhao
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Shi
- Department of Internal Medicine, Clinical Medicine School, Yunnan Traditional Chinese Medicine University, Kunming, China
| | - Qun-Yuan Zhao
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Sheng-Jie Sun
- Department of Emergency, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhen Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Si-Hang Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
18
|
Mahmoudi N, Wang Y, Moriarty N, Ahmed NY, Dehorter N, Lisowski L, Harvey AR, Parish CL, Williams RJ, Nisbet DR. Neuronal Replenishment via Hydrogel-Rationed Delivery of Reprogramming Factors. ACS NANO 2024; 18:3597-3613. [PMID: 38221746 DOI: 10.1021/acsnano.3c11337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The central nervous system's limited capacity for regeneration often leads to permanent neuronal loss following injury. Reprogramming resident reactive astrocytes into induced neurons at the site of injury is a promising strategy for neural repair, but challenges persist in stabilizing and accurately targeting viral vectors for transgene expression. In this study, we employed a bioinspired self-assembling peptide (SAP) hydrogel for the precise and controlled release of a hybrid adeno-associated virus (AAV) vector, AAVDJ, carrying the NeuroD1 neural reprogramming transgene. This method effectively mitigates the issues of high viral dosage at the target site, off-target delivery, and immunogenic reactions, enhancing the vector's targeting and reprogramming efficiency. In vitro, this vector successfully induced neuron formation, as confirmed by morphological, histochemical, and electrophysiological analyses. In vivo, SAP-mediated delivery of AAVDJ-NeuroD1 facilitated the trans-differentiation of reactive host astrocytes into induced neurons, concurrently reducing glial scarring. Our findings introduce a safe and effective method for treating central nervous system injuries, marking a significant advancement in regenerative neuroscience.
Collapse
Affiliation(s)
- Negar Mahmoudi
- Laboratory of Advanced Biomaterials, the John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
- ANU College of Engineering & Computer Science, Acton, ACT 2601, Australia
| | - Yi Wang
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Niamh Moriarty
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Noorya Y Ahmed
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nathalie Dehorter
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Vector and Genome Engineering Facility, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Australian Genome Therapeutics Centre, Children's Medical Research Institute and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, 04-141 Warsaw, Poland
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, and Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Richard J Williams
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- IMPACT, School of Medicine, Deakin University, Geelong, VIC 3217, Australia
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, the John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
19
|
Feng L, Chao J, Ye P, Luong Q, Sun G, Liu W, Cui Q, Flores S, Jackson N, Shayento ANH, Sun G, Liu Z, Hu W, Shi Y. Developing Hypoimmunogenic Human iPSC-Derived Oligodendrocyte Progenitor Cells as an Off-The-Shelf Cell Therapy for Myelin Disorders. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206910. [PMID: 37271923 PMCID: PMC10427412 DOI: 10.1002/advs.202206910] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Demyelinating disorders are among the most common and debilitating diseases in neurology. Canavan disease (CD) is a lethal demyelinating disease caused by mutation of the aspartoacylase (ASPA) gene, which leads to the accumulation of its substrate N-acetyl-l-aspartate (NAA), and consequently demyelination and vacuolation in the brain. In this study, hypoimmunogenic human induced pluripotent stem cell (iPSC)-derived oligodendrocyte progenitor cells (OPC) are developed from a healthy donor as an "off-the-shelf" cell therapy. Hypoimmunogenic iPSCs are generated through CRISPR/Cas9 editing of the human leukocyte antigen (HLA) molecules in healthy donor-derived iPSCs and differentiated into OPCs. The OPCs are engrafted into the brains of CD (nur7) mice and exhibit widespread distribution in the brain. The engrafted OPCs mature into oligodendrocytes that express the endogenous wildtype ASPA gene. Consequently, the transplanted mice exhibit elevated human ASPA expression and enzymatic activity and reduced NAA level in the brain. The transplanted OPCs are able to rescue major pathological features of CD, including defective myelination, extensive vacuolation, and motor function deficits. Moreover, the hypoimmunogenic OPCs exhibit low immunogenicity both in vitro and in vivo. The hypoimmunogenic OPCs can be used as "off-the-shelf" universal donor cells to treat various CD patients and many other demyelinating disorders, especially autoimmune demyelinating diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Lizhao Feng
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Jianfei Chao
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Peng Ye
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Qui Luong
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Guoqiang Sun
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Wei Liu
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Qi Cui
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Sergio Flores
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Natasha Jackson
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Afm Nazmul Hoque Shayento
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Guihua Sun
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Zhenqing Liu
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Weidong Hu
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
- Department of Immunology and TheranosticsBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| | - Yanhong Shi
- Department of Neurodegenerative DiseasesBeckman Research Institute of City of Hope1500 E. Duarte Rd.DuarteCA91010USA
| |
Collapse
|
20
|
Stogsdill JA, Harwell CC, Goldman SA. Astrocytes as master modulators of neural networks: Synaptic functions and disease-associated dysfunction of astrocytes. Ann N Y Acad Sci 2023; 1525:41-60. [PMID: 37219367 DOI: 10.1111/nyas.15004] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Astrocytes are the most abundant glial cell type in the central nervous system and are essential to the development, plasticity, and maintenance of neural circuits. Astrocytes are heterogeneous, with their diversity rooted in developmental programs modulated by the local brain environment. Astrocytes play integral roles in regulating and coordinating neural activity extending far beyond their metabolic support of neurons and other brain cell phenotypes. Both gray and white matter astrocytes occupy critical functional niches capable of modulating brain physiology on time scales slower than synaptic activity but faster than those adaptive responses requiring a structural change or adaptive myelination. Given their many associations and functional roles, it is not surprising that astrocytic dysfunction has been causally implicated in a broad set of neurodegenerative and neuropsychiatric disorders. In this review, we focus on recent discoveries concerning the contributions of astrocytes to the function of neural networks, with a dual focus on the contribution of astrocytes to synaptic development and maturation, and on their role in supporting myelin integrity, and hence conduction and its regulation. We then address the emerging roles of astrocytic dysfunction in disease pathogenesis and on potential strategies for targeting these cells for therapeutic purposes.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Steven A Goldman
- Sana Biotechnology Inc., Cambridge, Massachusetts, USA
- Center for Translational Neuromedicine, University of Rochester, Rochester, New York, USA
- University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
21
|
Coyoy-Salgado A, Orozco-Barrios C, Sánchez-Torres S, Olayo MG, Cruz GJ, Morales-Corona J, Olayo R, Diaz-Ruiz A, Ríos C, Alvarez-Mejia L, Mondragón-Lozano R, Morales-Guadarrama A, Alonso-García AL, Fabela-Sánchez O, Salgado-Ceballos H. Gene expression and locomotor recovery in adult rats with spinal cord injury and plasma-synthesized polypyrrole/iodine application combined with a mixed rehabilitation scheme. Front Neurol 2023; 14:1124245. [PMID: 37288064 PMCID: PMC10243140 DOI: 10.3389/fneur.2023.1124245] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/14/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Spinal cord injury (SCI) can cause paralysis, for which effective therapeutic strategies have not been developed yet. The only accepted strategy for patients is rehabilitation (RB), although this does not allow complete recovery of lost functions, which makes it necessary to combine it with strategies such as plasma-synthesized polypyrrole/iodine (PPy/I), a biopolymer with different physicochemical properties than PPy synthesized by conventional methods. After SCI in rats, PPy/I promotes functional recovery. Therefore, the purpose of this study was to increase the beneficial effects of both strategies and identify which genes activate PPy/I when applied alone or in combination with a mixed scheme of RB by swimming and enriched environment (SW/EE) in rats with SCI. Methods Microarray analysis was performed to identify mechanisms of action underlying the effects of PPy/I and PPy/I+SW/EE on motor function recovery as evaluated by the BBB scale. Results Results showed robust upregulation by PPy/I in genes related to the developmental process, biogenesis, synapse, and synaptic vesicle trafficking. In addition, PPy/I+SW/EE increased the expression of genes related to proliferation, biogenesis, cell development, morphogenesis, cell differentiation, neurogenesis, neuron development, and synapse formation processes. Immunofluorescence analysis showed the expression of β-III tubulin in all groups, a decreased expression of caspase-3 in the PPy/I group and GFAP in the PPy/I+SW/EE group (p < 0.05). Better preservation of nerve tissue was observed in PPy/I and PPy/SW/EE groups (p < 0.05). In the BBB scale, the control group scored 1.72 ± 0.41, animals with PPy/I treatment scored 4.23 ± 0.33, and those with PPy/I+SW/EE scored 9.13 ± 0.43 1 month after follow-up. Conclusion Thus, PPy/I+SW/EE could represent a therapeutic alternative for motor function recovery after SCI.
Collapse
Affiliation(s)
- Angélica Coyoy-Salgado
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Carlos Orozco-Barrios
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Stephanie Sánchez-Torres
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - María Guadalupe Olayo
- Instituto Nacional de Investigaciones Nucleares, Department of Physics, Axapusco, Mexico
| | - Guillermo Jesus Cruz
- Instituto Nacional de Investigaciones Nucleares, Department of Physics, Axapusco, Mexico
| | - Juan Morales-Corona
- Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Roberto Olayo
- Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Araceli Diaz-Ruiz
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez S.S.A., Department of Neurochemistry, Mexico City, Mexico
| | - Camilo Ríos
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez S.S.A., Department of Neurochemistry, Mexico City, Mexico
| | - Laura Alvarez-Mejia
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Rodrigo Mondragón-Lozano
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Axayacatl Morales-Guadarrama
- Electrical Engineering Department, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
- National Center for Research in Imaging and Medical Instrumentation, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | | | - Omar Fabela-Sánchez
- Researchers for Mexico CONACyT-Centro de Investigación en Química Aplicada, Department of Chemistry Macromolecules and Nanomaterials, Saltillo, Mexico
| | - Hermelinda Salgado-Ceballos
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| |
Collapse
|
22
|
Dai Y, Wang W, Zhou X, li L, Tang Y, Shao M, Lyu F. Biomimetic Electrospun PLLA/PPSB Nanofibrous Scaffold Combined with Human Neural Stem Cells for Spinal Cord Injury Repair. ACS APPLIED NANO MATERIALS 2023; 6:5980-5993. [DOI: 10.1021/acsanm.3c00374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Yuan Dai
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Weizhong Wang
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200240, China
| | - Xiaojun Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Linli li
- Department of Orthopedics, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| | - Yuyi Tang
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200240, China
| | - Minghao Shao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Feizhou Lyu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Orthopedics, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
23
|
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: 18] [Impact Index Per Article: 9.0] [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
|
24
|
Vieira S, Strymecka P, Stanaszek L, Silva-Correia J, Drela K, Fiedorowicz M, Malysz-Cymborska I, Janowski M, Reis RL, Łukomska B, Walczak P, Oliveira JM. Mn-Based Methacrylated Gellan Gum Hydrogels for MRI-Guided Cell Delivery and Imaging. Bioengineering (Basel) 2023; 10:bioengineering10040427. [PMID: 37106614 PMCID: PMC10135712 DOI: 10.3390/bioengineering10040427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
This work aims to engineer a new stable injectable Mn-based methacrylated gellan gum (Mn/GG-MA) hydrogel for real-time monitored cell delivery into the central nervous system. To enable the hydrogel visualization under Magnetic Resonance Imaging (MRI), GG-MA solutions were supplemented with paramagnetic Mn2+ ions before its ionic crosslink with artificial cerebrospinal fluid (aCSF). The resulting formulations were stable, detectable by T1-weighted MRI scans and also injectable. Cell-laden hydrogels were prepared using the Mn/GG-MA formulations, extruded into aCSF for crosslink, and after 7 days of culture, the encapsulated human adipose-derived stem cells remained viable, as assessed by Live/Dead assay. In vivo tests, using double mutant MBPshi/shi/rag2 immunocompromised mice, showed that the injection of Mn/GG-MA solutions resulted in a continuous and traceable hydrogel, visible on MRI scans. Summing up, the developed formulations are suitable for both non-invasive cell delivery techniques and image-guided neurointerventions, paving the way for new therapeutic procedures.
Collapse
Affiliation(s)
- Sílvia Vieira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Paulina Strymecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Joana Silva-Correia
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Katarzyna Drela
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Michał Fiedorowicz
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Izabela Malysz-Cymborska
- Department of Neurology and Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Rui Luís Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Barbara Łukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Piotr Walczak
- Department of Neurology and Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| | - Joaquim Miguel Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark—Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
- Correspondence: ; Tel.: +351-253510931; Fax: +351-253510909
| |
Collapse
|
25
|
Qian L, Yang K, Liu X, Zhang L, Zhao H, Qiu LZ, Chu Y, Hao W, Zhuang Y, Chen Y, Dai J. Baicalein-functionalized collagen scaffolds direct neuronal differentiation toward enhancing spinal cord injury repair. Biomater Sci 2023; 11:678-689. [PMID: 36511438 DOI: 10.1039/d2bm01467j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) repair remains a major challenge in clinics. Though neural stem cells (NSCs) have shown great potentials in SCI treatment, their applications were hampered since they primarily differentiate into astrocytes rather than neurons in the injured area, indicating a high demand for effective strategies to direct neuronal differentiation. Baicalein is a clinical drug with multiple pharmacological activities, while its effects on NSCs have rarely been reported. In the current work, inspired by a similarity of the metabolic reprogramming required in neuronal differentiation and that involved in chemoresistance reversal of cancer cells induced by baicalein, we studied the role of baicalein in NSC differentiation and discovered its promotion effects on neuronal differentiation. Based on this observation, baicalein-functionalized collagen scaffolds (BFCSs) were developed and applied for SCI treatment. The BFCSs released the payload in a sustained way and possessed comparable physical properties to the commonly used collagen. Both in vitro studies with primary NSCs and in vivo studies in SCI rats showed that the BFCSs containing a low amount of baicalein can facilitate not only neurogenesis and axon extension, but also reduce astrocyte production and glial scar formation. More importantly, the BFCS implantation led to improvement in the motor functional recovery of SCI rats. Thus, the BFCSs provided a potential strategy to induce neuronal differentiation towards facilitating SCI repair, as well as for the treatment of other central nervous system injuries.
Collapse
Affiliation(s)
- Lin Qian
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Keni Yang
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Xiru Liu
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Lulu Zhang
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Haitao Zhao
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Lin-Zi Qiu
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Yun Chu
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Wangping Hao
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Yan Zhuang
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Yanyan Chen
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China.
| | - Jianwu Dai
- Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123 China. .,Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
26
|
Carmichael ST, Llorente IL. The Ties That Bind: Glial Transplantation in White Matter Ischemia and Vascular Dementia. Neurotherapeutics 2023; 20:39-47. [PMID: 36357662 PMCID: PMC10119342 DOI: 10.1007/s13311-022-01322-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/12/2022] Open
Abstract
White matter injury is a progressive vascular disease that leads to neurological deficits and vascular dementia. It comprises up to 30% of all diagnosed strokes, though up to ten times as many events go undiagnosed in early stages. There are several pathologies that can lead to white matter injury. While some studies suggest that white matter injury starts as small infarcts in deep penetrating blood vessels in the brain, others point to the breakdown of endothelial function or the blood-brain barrier as the primary cause of the disease. Whether due to local endothelial or BBB dysfunction, or to local small infarcts (or a combination), white matter injury progresses, accumulates, and expands from preexisting lesions into adjacent white matter to produce motor and cognitive deficits that present as vascular dementia in the elderly. Vascular dementia is the second leading cause of dementia, and white matter injury-attributed vascular dementia represents 40% of all diagnosed dementias and aggravates Alzheimer's pathology. Despite the advances in the last 15 years, there are few animal models of progressive subcortical white matter injury or vascular dementia. This review will discuss recent progress in animal modeling of white matter injury and the emerging principles to enhance glial function as a means of promoting repair and recovery.
Collapse
Affiliation(s)
- S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E Young Drive South, NRB 407, Los Angeles, CA, 90095, USA
| | - Irene L Llorente
- Department of Neurosurgery, Stanford University, 3801 Miranda Ave, 94304, Palo alto, USA.
| |
Collapse
|
27
|
Becker J, Sun B, Alammari F, Haerty W, Vance KW, Szele FG. What has single-cell transcriptomics taught us about long non-coding RNAs in the ventricular-subventricular zone? Stem Cell Reports 2022; 18:354-376. [PMID: 36525965 PMCID: PMC9860170 DOI: 10.1016/j.stemcr.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNA (lncRNA) function is mediated by the process of transcription or through transcript-dependent associations with proteins or nucleic acids to control gene regulatory networks. Many lncRNAs are transcribed in the ventricular-subventricular zone (V-SVZ), a postnatal neural stem cell niche. lncRNAs in the V-SVZ are implicated in neurodevelopmental disorders, cancer, and brain disease, but their functions are poorly understood. V-SVZ neurogenesis capacity declines with age due to stem cell depletion and resistance to neural stem cell activation. Here we analyzed V-SVZ transcriptomics by pooling current single-cell RNA-seq data. They showed consistent lncRNA expression during stem cell activation, lineage progression, and aging. In conjunction with epigenetic and genetic data, we predicted V-SVZ lncRNAs that regulate stem cell activation and differentiation. Some of the lncRNAs validate known epigenetic mechanisms, but most remain uninvestigated. Our analysis points to several lncRNAs that likely participate in key aspects of V-SVZ stem cell activation and neurogenesis in health and disease.
Collapse
Affiliation(s)
- Jemima Becker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bin Sun
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Farah Alammari
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia,Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | - Keith W. Vance
- Department of Life Sciences, University of Bath, Bath, UK
| | - Francis George Szele
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
28
|
Rapid differentiation of hiPSCs into functional oligodendrocytes using an OLIG2 synthetic modified messenger RNA. Commun Biol 2022; 5:1095. [PMID: 36241911 PMCID: PMC9568531 DOI: 10.1038/s42003-022-04043-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/27/2022] [Indexed: 11/28/2022] Open
Abstract
Transcription factors (TFs) have been introduced to drive the highly efficient differentiation of human-induced pluripotent stem cells (hiPSCs) into lineage-specific oligodendrocytes (OLs). However, effective strategies currently rely mainly on genome-integrating viruses. Here we show that a synthetic modified messenger RNA (smRNA)-based reprogramming method that leads to the generation of transgene-free OLs has been developed. An smRNA encoding a modified form of OLIG2, in which the serine 147 phosphorylation site is replaced with alanine, OLIG2S147A, is designed to reprogram hiPSCs into OLs. We demonstrate that repeated administration of the smRNA encoding OLIG2S147A lead to higher and more stable protein expression. Using the single-mutant OLIG2 smRNA morphogen, we establish a 6-day smRNA transfection protocol, and glial induction lead to rapid NG2+ OL progenitor cell (OPC) generation (>70% purity) from hiPSC. The smRNA-induced NG2+ OPCs can mature into functional OLs in vitro and promote remyelination in vivo. Taken together, we present a safe and efficient smRNA-driven strategy for hiPSC differentiation into OLs, which may be utilized for therapeutic OPC/OL transplantation in patients with neurodegenerative disease. The use of synthetic modified messenger RNA (smRNA) allows for the differentiation of human-induced pluripotent stem cells (hiPSCs) into lineage-specific oligodendrocytes.
Collapse
|
29
|
Feng S, Zhang T, Ke W, Xiao Y, Guo Z, Lu C, Li S, Guo Z, Liu Y, Tan G, Chen Y, Yue F, Shu Y, Yue C, Jing N. The long-term survival and functional maturation of human iNPC-derived neurons in the basal forebrain of cynomolgus monkeys. LIFE MEDICINE 2022; 1:196-206. [PMID: 39871935 PMCID: PMC11749281 DOI: 10.1093/lifemedi/lnac008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/27/2022] [Indexed: 01/29/2025]
Abstract
Human induced neural stem/progenitor cells (iNPCs) are a promising source of cells for stem cell-based therapy. The therapeutic potential of human iNPCs has been extensively tested in animal models, including both mouse and monkey models. However, the comprehensive characterization of grafted iNPCs in the brain of non-human primates has been lagged behind. In this study, we transplanted human iNPCs into the basal forebrain of adult cynomolgus monkeys. We found that grafted iNPCs predominantly differentiated into neurons that displayed long-term survival up to 12 months. Additionally, iNPC-derived human neurons gradually matured in term of morphology and subtype differentiation. More excitingly, we observed that human neurons displayed electrophysiological activities resembling those of mature neurons, indicating the acquisition of functional membrane properties. Collectively, this study systematically characterized human iNPCs in the brain of non-human primates, and will provide invaluable clues for developing safe and effective stem cell-based therapies for different brain disorders.
Collapse
Affiliation(s)
- Su Feng
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou 510005, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- National Clinical Research Center for Ophthalmic Diseases, Shanghai 200080, China
| | - Wei Ke
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institutes for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yujie Xiao
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institutes for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Zhong Guo
- Wincon TheraCells Biotechnologies Co, LTD, Nanning 530000, China
| | - Chunling Lu
- Wincon TheraCells Biotechnologies Co, LTD, Nanning 530000, China
| | - Shuntang Li
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Zhongxin Guo
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yuanyuan Liu
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Guohe Tan
- Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Feng Yue
- Wincon TheraCells Biotechnologies Co, LTD, Nanning 530000, China
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Yousheng Shu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institutes for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Chunmei Yue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- Department of Biological Sciences, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou 215000, China
| | - Naihe Jing
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou 510005, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
30
|
Girão AF, Serrano MC, Completo A, Marques PAAP. Is Graphene Shortening the Path toward Spinal Cord Regeneration? ACS NANO 2022; 16:13430-13467. [PMID: 36000717 PMCID: PMC9776589 DOI: 10.1021/acsnano.2c04756] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Along with the development of the next generation of biomedical platforms, the inclusion of graphene-based materials (GBMs) into therapeutics for spinal cord injury (SCI) has potential to nourish topmost neuroprotective and neuroregenerative strategies for enhancing neural structural and physiological recovery. In the context of SCI, contemplated as one of the most convoluted challenges of modern medicine, this review first provides an overview of its characteristics and pathophysiological features. Then, the most relevant ongoing clinical trials targeting SCI, including pharmaceutical, robotics/neuromodulation, and scaffolding approaches, are introduced and discussed in sequence with the most important insights brought by GBMs into each particular topic. The current role of these nanomaterials on restoring the spinal cord microenvironment after injury is critically contextualized, while proposing future concepts and desirable outputs for graphene-based technologies aiming to reach clinical significance for SCI.
Collapse
Affiliation(s)
- André F. Girão
- Centre
for Mechanical Technology and Automation (TEMA), Department of Mechanical
Engineering, University of Aveiro (UA), Aveiro, 3810-193, Portugal
- Instituto
de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), Calle Sor Juana Inés de la
Cruz 3, Madrid, 28049, Spain
- (A.F.G.)
| | - María Concepcion Serrano
- Instituto
de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), Calle Sor Juana Inés de la
Cruz 3, Madrid, 28049, Spain
- (M.C.S.)
| | - António Completo
- Centre
for Mechanical Technology and Automation (TEMA), Department of Mechanical
Engineering, University of Aveiro (UA), Aveiro, 3810-193, Portugal
| | - Paula A. A. P. Marques
- Centre
for Mechanical Technology and Automation (TEMA), Department of Mechanical
Engineering, University of Aveiro (UA), Aveiro, 3810-193, Portugal
- (P.A.A.P.M.)
| |
Collapse
|
31
|
Yue C, Feng S, Chen Y, Jing N. The therapeutic prospects and challenges of human neural stem cells for the treatment of Alzheimer's Disease. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:28. [PMID: 36050613 PMCID: PMC9437172 DOI: 10.1186/s13619-022-00128-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder associated with aging. Due to its insidious onset, protracted progression, and unclear pathogenesis, it is considered one of the most obscure and intractable brain disorders, and currently, there are no effective therapies for it. Convincing evidence indicates that the irreversible decline of cognitive abilities in patients coincides with the deterioration and degeneration of neurons and synapses in the AD brain. Human neural stem cells (NSCs) hold the potential to functionally replace lost neurons, reinforce impaired synaptic networks, and repair the damaged AD brain. They have therefore received extensive attention as a possible source of donor cells for cellular replacement therapies for AD. Here, we review the progress in NSC-based transplantation studies in animal models of AD and assess the therapeutic advantages and challenges of human NSCs as donor cells. We then formulate a promising transplantation approach for the treatment of human AD, which would help to explore the disease-modifying cellular therapeutic strategy for the treatment of human AD.
Collapse
Affiliation(s)
- Chunmei Yue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China.
| | - Su Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou, 510005, China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Bioland Laboratory/Guangzhou Laboratory, Guangzhou, 510005, China.
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
32
|
McGinley LM, Chen KS, Mason SN, Rigan DM, Kwentus JF, Hayes JM, Glass ED, Reynolds EL, Murphy GG, Feldman EL. Monoclonal antibody-mediated immunosuppression enables long-term survival of transplanted human neural stem cells in mouse brain. Clin Transl Med 2022; 12:e1046. [PMID: 36101963 PMCID: PMC9471059 DOI: 10.1002/ctm2.1046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/14/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As the field of stem cell therapy advances, it is important to develop reliable methods to overcome host immune responses in animal models. This ensures survival of transplanted human stem cell grafts and enables predictive efficacy testing. Immunosuppressive drugs derived from clinical protocols are frequently used but are often inconsistent and associated with toxic side effects. Here, using a molecular imaging approach, we show that immunosuppression targeting costimulatory molecules CD4 and CD40L enables robust survival of human xenografts in mouse brain, as compared to conventional tacrolimus and mycophenolate mofetil. METHODS Human neural stem cells were modified to express green fluorescent protein and firefly luciferase. Cells were implanted in the fimbria fornix of the hippocampus and viability assessed by non-invasive bioluminescent imaging. Cell survival was assessed using traditional pharmacologic immunosuppression as compared to monoclonal antibodies directed against CD4 and CD40L. This paradigm was also implemented in a transgenic Alzheimer's disease mouse model. RESULTS Graft rejection occurs within 7 days in non-immunosuppressed mice and within 14 days in mice on a traditional regimen. The addition of dual monoclonal antibody immunosuppression extends graft survival past 7 weeks (p < .001) on initial studies. We confirm dual monoclonal antibody treatment is superior to either antibody alone (p < .001). Finally, we demonstrate robust xenograft survival at multiple cell doses up to 6 months in both C57BL/6J mice and a transgenic Alzheimer's disease model (p < .001). The dual monoclonal antibody protocol demonstrated no significant adverse effects, as determined by complete blood counts and toxicity screen. CONCLUSIONS This study demonstrates an effective immunosuppression protocol for preclinical testing of stem cell therapies. A transition towards antibody-based strategies may be advantageous by enabling stem cell survival in preclinical studies that could inform future clinical trials.
Collapse
Affiliation(s)
- Lisa M. McGinley
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin S. Chen
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Shayna N. Mason
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Diana M. Rigan
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - John M. Hayes
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Emily D. Glass
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Michigan Neuroscience InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Evan L. Reynolds
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Geoffrey G. Murphy
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Michigan Neuroscience InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
33
|
Chen X, Zhao T, Ke N, Qian Y, Wang W, Liu L, Liu C. In-vitro differentiation of human embryonic stem cells into spinal cord neural stem cells. Neuroreport 2022; 33:518-525. [PMID: 35882016 DOI: 10.1097/wnr.0000000000001812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In-vitro differentiation of human embryonic stem cells into spinal cord neural stem cells (NSCs) can help researchers better understand the cellular processes associated with spinal cord development and regeneration, and provide therapeutic strategies for spinal cord disorders. However, effective and consistent methods for the generation of human spinal cord NSCs are rare. Objective of the study is to establish methods for the in-vitro induction and long-term maintenance of human spinal cord NSCs. H9 cells were treated with neural induction medium for 10 days under single-cell seeding condition, followed by treatment with neural maintenance medium and replacement with NSC medium after five passages. The identity of the generated cells was determined by immunofluorescence, immunoblotting, and cleavage under targets and tagmentation (CUT&Tag) assays. After the neural induction, OCT4, an embryonic stem cell marker, was significantly reduced, whereas NESTIN and PAX6, two NSC markers, were clearly increased. After the neural maintenance, most of the H9-derived cells consistently expressed NESTIN and PAX6 together with SOX1 and HOXC9, two spinal cord markers. The Homer known motif enrichment results of the CUT&Tag assay confirmed the expression of HOXC9 in the H9-derived spinal cord NSCs, which can be maintained for more than 40 days under an in vitro culture system. This study sheds new light on effective induction and maintenance of human spinal cord NSCs.
Collapse
Affiliation(s)
- Xueying Chen
- School of Basic Medical Sciences, Anhui Medical University
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, Anhui Medical University
| | - Tianyi Zhao
- School of Basic Medical Sciences, Anhui Medical University
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, Anhui Medical University
| | - Naiyu Ke
- The First Clinical Medical College, Anhui Medical University
| | - Yutong Qian
- The First Clinical Medical College, Anhui Medical University
| | - Wanrong Wang
- The First Clinical Medical College, Anhui Medical University
| | - Lihua Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Chao Liu
- School of Basic Medical Sciences, Anhui Medical University
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, Anhui Medical University
| |
Collapse
|
34
|
Lu X, Lv C, Zhao Y, Wang Y, Li Y, Ji C, Wang Z, Ye W, Yu S, Bai J, Cai W. TSG-6 released from adipose stem cells-derived small extracellular vesicle protects against spinal cord ischemia reperfusion injury by inhibiting endoplasmic reticulum stress. Stem Cell Res Ther 2022; 13:291. [PMID: 35831906 PMCID: PMC9281104 DOI: 10.1186/s13287-022-02963-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/12/2022] [Indexed: 11/30/2022] Open
Abstract
Background Spinal cord ischemia reperfusion injury (SCIRI) is a complication of aortic aneurysm repair or spinal cord surgery that is associated with permanent neurological deficits. Mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEVs) have been shown to be potential therapeutic options for improving motor functions after SCIRI. Due to their easy access and multi-directional differentiation potential, adipose‐derived stem cells (ADSCs) are preferable for this application. However, the effects of ADSC-derived sEVs (ADSC-sEVs) on SCIRI have not been reported. Results We found that ADSC-sEVs inhibited SCIRI-induced neuronal apoptosis, degradation of tight junction proteins and suppressed endoplasmic reticulum (ER) stress. However, in the presence of the ER stress inducer, tunicamycin, its anti-apoptotic and blood–spinal cord barrier (BSCB) protective effects were significantly reversed. We found that ADSC-sEVs contain tumor necrosis factor (TNF)-stimulated gene-6 (TSG-6) whose overexpression inhibited ER stress in vivo by modulating the PI3K/AKT pathway. Conclusions ADSC-sEVs inhibit neuronal apoptosis and BSCB disruption in SCIRI by transmitting TSG-6, which suppresses ER stress by modulating the PI3K/AKT pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02963-4.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China.,Department of Orthopaedics, Dongtai Hospital Affiliated to Nantong University, Dongtai City, Jiangsu, China
| | - Chengtang Lv
- Department of Orthopaedics, Yancheng Third People's Hospital, Yancheng, 224000, Jiangsu, China
| | - Yuechao Zhao
- Department of Orthopedic Oncology, Changzheng Hospital, Secondary Military Medical University, Shanghai, China.,Department of Orthopedic, PLA Navy No.905 Hospital, Secondary Military Medical University, Shanghai, China
| | - Yufei Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Haining, Zhejiang, China
| | - Yao Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, China
| | - Chengyue Ji
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Zhuanghui Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Wu Ye
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Shunzhi Yu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, China.
| | - Jianling Bai
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Jiangsu Province, Nanjing, 211166, China.
| | - Weihua Cai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, China.
| |
Collapse
|
35
|
Wang J, Chen S, Pan C, Li G, Tang Z. Application of Small Molecules in the Central Nervous System Direct Neuronal Reprogramming. Front Bioeng Biotechnol 2022; 10:799152. [PMID: 35875485 PMCID: PMC9301571 DOI: 10.3389/fbioe.2022.799152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of regenerative capacity of neurons leads to poor prognoses for some neurological disorders. The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases. In this review, the mechanisms of action of different small molecules, the approaches to screening small molecule cocktails, and the methods employed to detect their reprogramming efficiency are discussed, and the studies, focusing on neuronal reprogramming using small molecules in neurological disease models, are collected. Future research efforts are needed to investigate the in vivo mechanisms of small molecule-mediated neuronal reprogramming under pathophysiological states, optimize screening cocktails and dosing regimens, and identify safe and effective delivery routes to promote neural regeneration in different neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Gaigai Li
- *Correspondence: Gaigai Li, ; Zhouping Tang,
| | | |
Collapse
|
36
|
2D Ti 3C 2T xMXene couples electrical stimulation to promote proliferation and neural differentiation of neural stem cells. Acta Biomater 2022; 139:105-117. [PMID: 33348061 DOI: 10.1016/j.actbio.2020.12.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 01/31/2023]
Abstract
Preclinical studies involving stem cells require efficient physiochemical regulations on the fate of such cells. Because of their unique planar structure, metallic conductivity, and flexible surface functionalization, MXenes show potential for modulating stem cell fate. Here, the Ti3C2TxMXenenanosheets are dispersed on tissue culture polystyrene (TCPS). When primary mouse neural stem cells (NSCs) are cultured on laminin-coated Ti3C2TxMXene film, they form stable adhesion, retain their proliferative ability, and show extensive spreading of terminal extensions. With respect to their functional activity, NSCs cultured on Ti3C2TxMXene films form more active and synchronous network activity than those cultured on TCPS substrates. Moreover, Ti3C2TxMXene film significantly promotes the neural differentiation and the neurons have longer neurites and greater numbers of branch points and branch tips. NSC-derived neurons grown on the Ti3C2Tx MXene film preserved normal synapse development. Finally, electrical stimulation coupled with Ti3C2TxMXene film significantly enhances the proliferation of NSCs. These results indicate that Ti3C2TxMXene is an efficient interface for the proliferation and neural differentiation of NSC and the maturation of NSC-derived neurons, which expands the potential uses of the MXene family of materials and provides new strategies for stem cell studies. STATEMENT OF SIGNIFICANCE: The 2DTi3C2TxMXenenanosheets were applied to be an interface for regulating neural stem cells (NSCs). NSCs cultured on Ti3C2TxMXene film possessed higher proliferative ability with higher and more synchronous electrical activities. Moreover, Ti3C2TxMXene film significantly promoted the neural differentiation ratio of NSCs, and the neurons derived from NSCs cultured on Ti3C2TxMXene film had longer neurites and greater numbers of branch points and branch tips.When electrical stimulation was applied to NSCs via the Ti3C2TxMXene film, it significantly enhanced the proliferation of NSCs. This work expands the potential uses of the MXene family of materials and provides new strategies for stem cell studies.
Collapse
|
37
|
Yang P, Cao Q, Liu Y, Wang K, Zhu W. Small‐molecule‐driven direct reprogramming of Müller cells into bipolar‐like cells. Cell Prolif 2022; 55:e13184. [PMID: 35043487 PMCID: PMC8828256 DOI: 10.1111/cpr.13184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Pang Yang
- Department of Pharmacology School of Pharmacy Qingdao University Qingdao China
| | - Qilong Cao
- Qingdao Haier Biotech Co. Ltd Qingdao China
| | - Yani Liu
- Department of Pharmacology School of Pharmacy Qingdao University Qingdao China
| | - KeWei Wang
- Department of Pharmacology School of Pharmacy Qingdao University Qingdao China
- Institute of Innovative Drugs Qingdao University Qingdao China
| | - Wei Zhu
- Department of Pharmacology School of Pharmacy Qingdao University Qingdao China
- Shenzhen Ruipuxun Academy for Stem Cell & Regenerative Medicine Shen Zhen China
- Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine Beihang University & Capital Medical University Beijing China
| |
Collapse
|
38
|
Nouri P, Zimmer A, Brüggemann S, Friedrich R, Kühn R, Prakash N. Generation of a NES-mScarlet Red Fluorescent Reporter Human iPSC Line for Live Cell Imaging and Flow Cytometric Analysis and Sorting Using CRISPR-Cas9-Mediated Gene Editing. Cells 2022; 11:268. [PMID: 35053384 PMCID: PMC8773741 DOI: 10.3390/cells11020268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/16/2022] Open
Abstract
Advances in the regenerative stem cell field have propelled the generation of tissue-specific cells in the culture dish for subsequent transplantation, drug screening purposes, or the elucidation of disease mechanisms. One major obstacle is the heterogeneity of these cultures, in which the tissue-specific cells of interest usually represent only a fraction of all generated cells. Direct identification of the cells of interest and the ability to specifically isolate these cells in vitro is, thus, highly desirable for these applications. The type VI intermediate filament protein NESTIN is widely used as a marker for neural stem/progenitor cells (NSCs/NPCs) in the developing and adult central and peripheral nervous systems. Applying CRISPR-Cas9 technology, we have introduced a red fluorescent reporter (mScarlet) into the NESTIN (NES) locus of a human induced pluripotent stem cell (hiPSC) line. We describe the generation and characterization of NES-mScarlet reporter hiPSCs and demonstrate that this line is an accurate reporter of NSCs/NPCs during their directed differentiation into human midbrain dopaminergic (mDA) neurons. Furthermore, NES-mScarlet hiPSCs can be used for direct identification during live cell imaging and for flow cytometric analysis and sorting of red fluorescent NSCs/NPCs in this paradigm.
Collapse
Affiliation(s)
- Parivash Nouri
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| | - Anja Zimmer
- Genome Engineering & Disease Models, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
| | - Stefanie Brüggemann
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| | - Robin Friedrich
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| | - Ralf Kühn
- Genome Engineering & Disease Models, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany;
| | - Nilima Prakash
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany; (P.N.); (S.B.); (R.F.)
| |
Collapse
|
39
|
Chand K, Nano R, Wixey J, Patel J. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:372-382. [PMID: 35485440 PMCID: PMC9052430 DOI: 10.1093/stcltm/szac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/12/2021] [Indexed: 11/25/2022] Open
Abstract
Fetal growth restriction (FGR) occurs when a fetus is unable to grow normally due to inadequate nutrient and oxygen supply from the placenta. Children born with FGR are at high risk of lifelong adverse neurodevelopmental outcomes, such as cerebral palsy, behavioral issues, and learning and attention difficulties. Unfortunately, there is no treatment to protect the FGR newborn from these adverse neurological outcomes. Chronic inflammation and vascular disruption are prevalent in the brains of FGR neonates and therefore targeted treatments may be key to neuroprotection. Tissue repair and regeneration via stem cell therapies have emerged as a potential clinical intervention for FGR babies at risk for neurological impairment and long-term disability. This review discusses the advancement of research into stem cell therapy for treating neurological diseases and how this may be extended for use in the FGR newborn. Leading preclinical studies using stem cell therapies in FGR animal models will be highlighted and the near-term steps that need to be taken for the development of future clinical trials.
Collapse
Affiliation(s)
- Kirat Chand
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Rachel Nano
- Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Julie Wixey
- Julie Wixey, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland Centre for Clinical Research, Herston 4029 QLD, Australia.
| | - Jatin Patel
- Corresponding authors: Jatin Patel, Translational Research Institute, Queensland University of Technology, 37 Kent Street, Woolloongabba 4102 QLD, Australia.
| |
Collapse
|
40
|
Proteomic Analysis of Human Neural Stem Cell Differentiation by SWATH-MS. Methods Mol Biol 2022; 2520:335-360. [PMID: 35579839 DOI: 10.1007/7651_2022_462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The unique properties of stem cells to self-renew and differentiate hold great promise in disease modelling and regenerative medicine. However, more information about basic stem cell biology and thorough characterization of available stem cell lines is needed. This is especially essential to ensure safety before any possible clinical use of stem cells or partially committed cell lines. As proteins are the key effector molecules in the cell, the proteomic characterization of cell lines, cell compartments or cell secretome and microenvironment is highly beneficial to answer above mentioned questions. Nowadays, method of choice for large-scale discovery-based proteomic analysis is mass spectrometry (MS) with data-independent acquisition (DIA). DIA is a robust, highly reproducible, high-throughput quantitative MS approach that enables relative quantification of thousands of proteins in one sample. In the current protocol, we describe a specific variant of DIA known as SWATH-MS for characterization of neural stem cell differentiation. The protocol covers the whole process from cell culture, sample preparation for MS analysis, the SWATH-MS data acquisition on TTOF 5600, the complete SWATH-MS data processing and quality control using Skyline software and the basic statistical analysis in R and MSstats package. The protocol for SWATH-MS data acquisition and analysis can be easily adapted to other samples amenable to MS-based proteomics.
Collapse
|
41
|
Feng Y, Bai S, Li G, Nie H, Chen S, Pan C, Zhang P, Tang Y, Liu N, Tang Z. Reprogramming rat astrocytes into neurons using small molecules for cell replacement following intracerebral hemorrhage. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Astrocytes are promising source cells to replace neurons lost to disease owing to a shared lineage and capacities for dedifferentiation and proliferation under pathological conditions. Reprogramming of astrocytes to neurons has been achieved by transcription factor modulation, but reprogramming in vitro or in vivo using small‐molecule drugs may have several advantages for clinical application. For instance, small molecules can be extensively characterized for efficacy, toxicity, and tumorigenicity in vitro; induce rapid initiation and subsequent reversal of transdifferentiation upon withdrawal, and obviate the need for exogenous gene transfection. Here we report a new astrocyte–neuron reprogramming strategy using a combination of small molecules (0.5 mM valproic acid, 1 μM RepSox, 3 μM CHIR99021, 2 μM I‐BET151, 10 μM ISX‐9, and 10 μM forskolin). Treatment with this drug combination gradually reduced expression levels of astroglial marker proteins (glial fibrillary acidic protein and S100), transiently enhanced expression of the neuronal progenitor marker doublecortin, and subsequently elevated expression of the mature neuronal marker NeuN in primary astrocyte cultures. These changes were accompanied by transition to a neuron‐like morphological phenotype and expression of multiple neuronal transcription factors. Further, this drug combination induced astrocyte‐to‐neuron transdifferentiation in a culture model of intracerebral hemorrhage (ICH) and upregulated many transdifferentiation‐associated signaling molecules in ICH model rats. In culture, the drug combination also reduced ICH model‐associated oxidative stress, apoptosis, and pro‐inflammatory cytokine production. Neurons derived from small‐molecule reprogramming of astrocytes in adult Sprague–Dawley rats demonstrated long‐term survival and maintenance of neuronal phenotype. This small‐molecule‐induced astrocyte‐to‐neuron transdifferentiation method may be a promising strategy for neuronal replacement therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Na Liu
- These authors contributed equally to this work
| | | |
Collapse
|
42
|
Motavaf M, Piao X. Oligodendrocyte Development and Implication in Perinatal White Matter Injury. Front Cell Neurosci 2021; 15:764486. [PMID: 34803612 PMCID: PMC8599582 DOI: 10.3389/fncel.2021.764486] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Perinatal white matter injury (WMI) is the most common brain injury in premature infants and can lead to life-long neurological deficits such as cerebral palsy. Preterm birth is typically accompanied by inflammation and hypoxic-ischemic events. Such perinatal insults negatively impact maturation of oligodendrocytes (OLs) and cause myelination failure. At present, no treatment options are clinically available to prevent or cure WMI. Given that arrested OL maturation plays a central role in the etiology of perinatal WMI, an increased interest has emerged regarding the functional restoration of these cells as potential therapeutic strategy. Cell transplantation and promoting endogenous oligodendrocyte function are two potential options to address this major unmet need. In this review, we highlight the underlying pathophysiology of WMI with a specific focus on OL biology and their implication for the development of new therapeutic targets.
Collapse
Affiliation(s)
- Mahsa Motavaf
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Xianhua Piao
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,Newborn Brain Research Institute, University of California, San Francisco, San Francisco, CA, United States.,Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, United States.,Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
43
|
Rathnam C, Yang L, Castro-Pedrido S, Luo J, Cai L, Lee KB. Hybrid SMART spheroids to enhance stem cell therapy for CNS injuries. SCIENCE ADVANCES 2021; 7:eabj2281. [PMID: 34586845 PMCID: PMC8480929 DOI: 10.1126/sciadv.abj2281] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Although stem cell therapy holds enormous potential for treating debilitating injuries and diseases in the central nervous system, low survival and inefficient differentiation have restricted its clinical applications. Recently, 3D cell culture methods, such as stem cell–based spheroids and organoids, have demonstrated advantages by incorporating tissue-mimetic 3D cell-cell interactions. However, a lack of drug and nutrient diffusion, insufficient cell-matrix interactions, and tedious fabrication procedures have compromised their therapeutic effects in vivo. To address these issues, we developed a biodegradable nanomaterial-templated 3D cell assembly method that enables the formation of hybrid stem cell spheroids with deep drug delivery capabilities and homogeneous incorporation of 3D cell-matrix interactions. Hence, high survival rates, controlled differentiation, and functional recovery were demonstrated in a spinal cord injury animal model. Overall, our hybrid stem cell spheroids represent a substantial development of material-facilitated 3D cell culture systems and can pave the way for stem cell–based treatment of CNS injuries.
Collapse
Affiliation(s)
- Christopher Rathnam
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sofia Castro-Pedrido
- Department of Biomedical Engineering Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jeffrey Luo
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Li Cai
- Department of Biomedical Engineering Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
44
|
Zou T, Jiang S, Yi B, Chen Q, Heng BC, Zhang C. Gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor enhances small molecule-induced neurogenic differentiation of stem cells from apical papilla. J Biomed Mater Res A 2021; 110:623-634. [PMID: 34590393 DOI: 10.1002/jbm.a.37315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
The limited neurogenic potential of adult stem cells and their non-specific lineage differentiation pose major challenges in cell-replacement therapy for neurological disorders. In our previous study, we demonstrated that the neurogenic potential of stem cells from apical papilla (SCAPs) was significantly improved upon induction with a small molecule cocktail. This study attempted to investigate whether neuronal differentiation of SCAPs induced by a small molecule cocktail can be further enhanced in a three-dimensional gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor (BDNF-GelMA). The physiological properties and neural differentiation of SCAPs treated with a combination of small molecules and BDNF-GelMA were evaluated by CCK8, Live/Dead assay, quantitative reverse transcription-polymerase chain reaction, western blot and immunocytochemistry. SCAPs embedded in BDNF-GelMA displayed superior morphological characteristics when induced by a small molecule cocktail, similar to neuronal phenotypes as compared to pure GelMA. There was significant upregulation of neural markers including Tuj1 and MAP2 by SCAPs embedded in BDNF-GelMA, as compared to pure GelMA. Hence, GelMA hydrogel loaded with a potent neurotrophic factor (BDNF) provides a conducive scaffold that can further enhance the differentiation of small molecule-treated SCAPs into neuronal-like cells, which may provide a therapeutic platform for the management of neurological disorders.
Collapse
Affiliation(s)
- Ting Zou
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shan Jiang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Baicheng Yi
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qixin Chen
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | | | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
45
|
Stem cells from human exfoliated deciduous teeth affect mitochondria and reverse cognitive decline in a senescence-accelerated mouse prone 8 model. Cytotherapy 2021; 24:59-71. [PMID: 34598900 DOI: 10.1016/j.jcyt.2021.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/31/2021] [Accepted: 07/31/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND AIMS Stem cell therapy is a novel therapy being explored for AD. The molecular mechanism of its effect is still unclear. The authors investigated the effects and mechanism by injection of SHEDs into an AD mouse model. METHODS SHEDs were cultured in vitro and injected into AD SAMP8 mice by caudal vein, and SHEDs labeled via synthetic dye showed in vivo migration to the head. The cognitive ability of SAMP8 mice was evaluated via Barnes maze and new object recognition. The pathological indicators of AD, including Tau, amyloid plaques and inflammatory factors, were examined at the protein or RNA level. Next, macro-proteomics analysis and weighted gene co-expression network analysis (WGCNA) based on protein groups and behavioral data were applied to discover the important gene cluster involved in the improvement of AD by SHEDs, which was further confirmed in an AD model in both mouse and cell lines. RESULTS SHED treatment improved the cognitive ability and pathological symptoms of SAMP8 mice. Proteomics analysis indicated that these improvements were tightly related to the mitochondria, which was proved through examination of the shape and function of mitochondria both in vivo (SAMP8 brain) and in vitro (SH-SY5Y cells). Finally, the core targets of SHEDs in the mitochondrial pathway, Hook3, Mic13 and MIF, were screened out and confirmed in vivo. CONCLUSIONS SHED treatment significantly relieved AD symptoms, improved cognitive ability and reversed memory loss in an AD mouse model, possibly through the recovery of dysfunctional mitochondria. These results raise the possibility that SHED may ease the symptoms of AD by targeting the mitochondria.
Collapse
|
46
|
Salidroside-pretreated mesenchymal stem cells contribute to neuroprotection in cerebral ischemic injury in vitro and in vivo. J Mol Histol 2021; 52:1145-1154. [PMID: 34570327 DOI: 10.1007/s10735-021-10022-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/23/2021] [Indexed: 01/19/2023]
Abstract
Mesenchymal stem cells (MSCs) are considered a promising tool for treating cerebral ischemic injury. However, their poor survival after transplantation limits their therapeutic effect and applications. Salidroside has been reported to exert potent cytoprotective and neuroprotective effects. This study aimed to investigate whether salidroside could improve MSC survival under hypoxic-ischemic conditions and, subsequently, alleviate cerebral ischemic injury in a rat model. MSCs were pretreated by salidroside under hypoxic-ischemic conditions. The cell proliferation, migratory capacity, and apoptosis were evaluated by means of Cell Counting Kit-8, transwell assay, and flow cytometry. MSCs pretreated with salidroside were transplanted into the rats subsequent to middle cerebral artery occlusion. The grip strength, 2,3,5-triphenyltetrazolium chloride, and hematoxylin-eosin staining were used to analyze the therapeutic efficiency and pathological changes. The mature neuron marker NeuN and astrocyte marker GFAP in the focal area were detected by immunofluorescence. These results indicated that salidroside promoted the proliferation, migration and reduced apoptosis of MSCs under hypoxic-ischemic conditions. In vivo experiments revealed that transplantation of salidroside-pretreated MSCs strengthened the therapeutic efficiency by enhancing neurogenesis and inhibiting neuroinflammation in the hippocampal CA1 area after ischemia. Our results suggest that pretreatment with salidroside could be an effective strategy to enhance the cell survival rate and the therapeutic effect of MSCs in treating cerebral ischemic injury.
Collapse
|
47
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
Collapse
|
48
|
Derivation of Oligodendrocyte Precursors from Adult Bone Marrow Stromal Cells for Remyelination Therapy. Cells 2021; 10:cells10082166. [PMID: 34440935 PMCID: PMC8391516 DOI: 10.3390/cells10082166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 01/04/2023] Open
Abstract
Transplantation of oligodendrocyte precursors (OPs) is potentially therapeutic for myelin disorders but a safe and accessible cell source remains to be identified. Here we report a two-step protocol for derivation of highly enriched populations of OPs from bone marrow stromal cells of young adult rats (aMSCs). Neural progenitors among the aMSCs were expanded in non-adherent sphere-forming cultures and subsequently directed along the OP lineage with the use of glial-inducing growth factors. Immunocytochemical and flow cytometric analyses of these cells confirmed OP-like expression of Olig2, PDGFRα, NG2, and Sox10. OPs so derived formed compact myelin both in vitro, as in co-culture with purified neurons, and in vivo, following transplantation into the corpus callosum of neonatal shiverer mice. Not only did the density of myelinated axons in the corpus callosum of recipient shiverer mice reach levels comparable to those in age-matched wild-type mice, but the mean lifespan of recipient shiverer mice also far exceeded those of non-recipient shiverer mice. Our results thus promise progress in harnessing the OP-generating potential of aMSCs towards cell therapy for myelin disorders.
Collapse
|
49
|
Marymonchyk A, Malvaut S, Saghatelyan A. In vivo live imaging of postnatal neural stem cells. Development 2021; 148:271820. [PMID: 34383894 DOI: 10.1242/dev.199778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neural stem cells (NSCs) are maintained in specific regions of the postnatal brain and contribute to its structural and functional plasticity. However, the long-term renewal potential of NSCs and their mode of division remain elusive. The use of advanced in vivo live imaging approaches may expand our knowledge of NSC physiology and provide new information for cell replacement therapies. In this Review, we discuss the in vivo imaging methods used to study NSC dynamics and recent live-imaging results with respect to specific intracellular pathways that allow NSCs to integrate and decode different micro-environmental signals. Lastly, we discuss future directions that may provide answers to unresolved questions regarding NSC physiology.
Collapse
Affiliation(s)
- Alina Marymonchyk
- CERVO Brain Research Center, Quebec City, QC, CanadaG1J 2G3.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, CanadaG1V 0A6
| | - Sarah Malvaut
- CERVO Brain Research Center, Quebec City, QC, CanadaG1J 2G3.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, CanadaG1V 0A6
| | - Armen Saghatelyan
- CERVO Brain Research Center, Quebec City, QC, CanadaG1J 2G3.,Department of Psychiatry and Neuroscience, Université Laval, Quebec City, QC, CanadaG1V 0A6
| |
Collapse
|
50
|
Wang F, Cheng L, Zhang X. Reprogramming Glial Cells into Functional Neurons for Neuro-regeneration: Challenges and Promise. Neurosci Bull 2021; 37:1625-1636. [PMID: 34283396 DOI: 10.1007/s12264-021-00751-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/24/2021] [Indexed: 01/02/2023] Open
Abstract
The capacity for neurogenesis in the adult mammalian brain is extremely limited and highly restricted to a few regions, which greatly hampers neuronal regeneration and functional restoration after neuronal loss caused by injury or disease. Meanwhile, transplantation of exogenous neuronal stem cells into the brain encounters several serious issues including immune rejection and the risk of tumorigenesis. Recent discoveries of direct reprogramming of endogenous glial cells into functional neurons have provided new opportunities for adult neuro-regeneration. Here, we extensively review the experimental findings of the direct conversion of glial cells to neurons in vitro and in vivo and discuss the remaining issues and challenges related to the glial subtypes and the specificity and efficiency of direct cell-reprograming, as well as the influence of the microenvironment. Although in situ glial cell reprogramming offers great potential for neuronal repair in the injured or diseased brain, it still needs a large amount of research to pave the way to therapeutic application.
Collapse
Affiliation(s)
- Fengchao Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Leping Cheng
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, and Guangxi Key Laboratory of Regenerative Medicine, Center for Translational Medicine, Guangxi Medical University, Nanning, 530021, China. .,Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China. .,Guangxi Health Commission Key Laboratory of Basic Research on Brain Function and Disease, Guangxi Medical University, Nanning, 530021, China.
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|