1
|
Ruggiero M, Cianciulli A, Calvello R, Lofrumento DD, Saponaro C, Filannino FM, Porro C, Panaro MA. Lactoferrin Attenuates Pro-Inflammatory Response and Promotes the Conversion into Neuronal Lineages in the Astrocytes. Int J Mol Sci 2025; 26:405. [PMID: 39796258 PMCID: PMC11720426 DOI: 10.3390/ijms26010405] [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: 11/22/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Neurodegenerative diseases are characterized by progressive loss of neurons and persistent inflammation. Neurons are terminally differentiated cells, and lost neurons cannot be replaced since neurogenesis is restricted to only two neurogenic niches in the adult brain, whose neurogenic potential decreases with age. In this regard, the astrocytes reprogramming into neurons may represent a promising strategy for restoring the lost neurons and rebuilding neural circuits. To date, many anti-inflammatory agents have been shown to reduce neuroinflammation; however, their potential to restore neuronal loss was poorly investigated. This study investigates the anti-inflammatory effects of lactoferrin on DI-TNC1 astrocyte cell line and its ability to induce astrocyte reprogramming in a context of sustained inflammation. For this purpose, astrocytes were pre-treated with lactoferrin (4 μg/mL) for 24 h, then with lipopolysaccharide (LPS) (400 ng/mL), and examined 2, 9 and 16 days from treatment. The results demonstrate that lactoferrin attenuates astrocyte reactivity by reducing Toll-like receptor 4 (TLR4), Glial fibrillary acidic protein (GFAP) and IL-6 expression, as well as by upregulating Interleukin-10 (IL-10) cytokine and NRF2 expression. Moreover, lactoferrin promotes the reprogramming of reactive astrocytes into proliferative neuroblasts by inducing the overexpression of the Sex determining region Y/SRY-box 2 (SOX2) reprogramming transcription factor. Overall, this study highlights the potential effects of lactoferrin to attenuate neuroinflammation and improve neurogenesis, suggesting a future strategy for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Melania Ruggiero
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, 73100 Lecce, Italy;
| | - Concetta Saponaro
- IRCCS Istituto Tumori Giovanni Paolo II, V.le O. Flacco, 65, 70124 Bari, Italy;
| | - Francesca Martina Filannino
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (F.M.F.); (C.P.)
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (F.M.F.); (C.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| |
Collapse
|
2
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Dong W, Liu S, Li S, Wang Z. Cell reprogramming therapy for Parkinson's disease. Neural Regen Res 2024; 19:2444-2455. [PMID: 38526281 PMCID: PMC11090434 DOI: 10.4103/1673-5374.390965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/23/2023] [Accepted: 10/08/2023] [Indexed: 03/26/2024] Open
Abstract
Parkinson's disease is typically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Many studies have been performed based on the supplementation of lost dopaminergic neurons to treat Parkinson's disease. The initial strategy for cell replacement therapy used human fetal ventral midbrain and human embryonic stem cells to treat Parkinson's disease, which could substantially alleviate the symptoms of Parkinson's disease in clinical practice. However, ethical issues and tumor formation were limitations of its clinical application. Induced pluripotent stem cells can be acquired without sacrificing human embryos, which eliminates the huge ethical barriers of human stem cell therapy. Another widely considered neuronal regeneration strategy is to directly reprogram fibroblasts and astrocytes into neurons, without the need for intermediate proliferation states, thus avoiding issues of immune rejection and tumor formation. Both induced pluripotent stem cells and direct reprogramming of lineage cells have shown promising results in the treatment of Parkinson's disease. However, there are also ethical concerns and the risk of tumor formation that need to be addressed. This review highlights the current application status of cell reprogramming in the treatment of Parkinson's disease, focusing on the use of induced pluripotent stem cells in cell replacement therapy, including preclinical animal models and progress in clinical research. The review also discusses the advancements in direct reprogramming of lineage cells in the treatment of Parkinson's disease, as well as the controversy surrounding in vivo reprogramming. These findings suggest that cell reprogramming may hold great promise as a potential strategy for treating Parkinson's disease.
Collapse
Affiliation(s)
- Wenjing Dong
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shuyi Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| |
Collapse
|
4
|
Parmar B, Bhatia D. Small Molecular Approaches for Cellular Reprogramming and Tissue Engineering: Functions as Mediators of the Cell Signaling Pathway. Biochemistry 2024; 63:2542-2556. [PMID: 39312802 DOI: 10.1021/acs.biochem.4c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Utilizing induced pluripotent stem cells (iPSCs) in drug screening and cell replacement therapy has emerged as a method with revolutionary applications. With the advent of patient-specific iPSCs and the subsequent development of cells that exhibit disease phenotypes, the focus of medication research will now shift toward the pathology of human diseases. Regular iPSCs can also be utilized to generate cells that assess the negative impacts of medications. These cells provide a much more precise and cost-efficient approach compared to many animal models. In this review, we explore the utilization of small-molecule drugs to enhance the growth of iPSCs and gain insights into the process of reprogramming. We mainly focus on the functions of small molecules in modulating different signaling pathways, thereby modulating cell fate. Understanding the way small molecule drugs interact with iPSC technology has the potential to significantly enhance the understanding of physiological pathways in stem cells and practical applications of iPSC-based therapy and screening systems, revolutionizing the treatment of diseases.
Collapse
Affiliation(s)
- Bhagyesh Parmar
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Palaj, Gandhinagar 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Palaj, Gandhinagar 382355, India
| |
Collapse
|
5
|
Li ST, Wan Y, Chen L, Ding Y. Advances in neuronal reprogramming for neurodegenerative diseases: Strategies, controversies, and opportunities. Exp Neurol 2024; 378:114817. [PMID: 38763354 DOI: 10.1016/j.expneurol.2024.114817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Neuronal death is often observed in central nervous system injuries and neurodegenerative diseases. The mammalian central nervous system manifests limited neuronal regeneration capabilities, and traditional cell therapies are limited in their potential applications due to finite cell sources and immune rejection. Neuronal reprogramming has emerged as a novel technology, in which non-neuronal cells (e.g. glial cells) are transdifferentiated into mature neurons. This process results in relatively minimal immune rejection. The present review discuss the latest progress in this cutting-edge field, including starter cell selection, innovative technical strategies and methods of neuronal reprogramming for neurodegenerative diseases, as well as the potential problems and controversies. The further development of neuronal reprogramming technology may pave the way for novel therapeutic strategies in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Si-Tong Li
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yue Wan
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yan Ding
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Wei J, Wang M, Li S, Han R, Xu W, Zhao A, Yu Q, Li H, Li M, Chi G. Reprogramming of astrocytes and glioma cells into neurons for central nervous system repair and glioblastoma therapy. Biomed Pharmacother 2024; 176:116806. [PMID: 38796971 DOI: 10.1016/j.biopha.2024.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.
Collapse
Affiliation(s)
- Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Shilin Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue, Changchun, Jilin Province 130021, China.
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
7
|
Xie X, Liu J. New role of astrocytes in neuroprotective mechanisms after ischemic stroke. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:748-755. [PMID: 37647906 PMCID: PMC10468254 DOI: 10.1055/s-0043-1770352] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/15/2023] [Indexed: 09/01/2023]
Abstract
Astrocytes are the most abundant cell subtypes in the central nervous system. Previous studies believed that astrocytes are supporting cells in the brain, which only provide nutrients for neurons. However, recent studies have found that astrocytes have more crucial and complex functions in the brain, such as neurogenesis, phagocytosis, and ischemic tolerance. After an ischemic stroke, the activated astrocytes can exert neuroprotective or neurotoxic effects through a variety of pathways. In this review, we will discuss the neuroprotective mechanisms of astrocytes in cerebral ischemia, and mainly focus on reactive astrocytosis or glial scar, neurogenesis, phagocytosis, and cerebral ischemic tolerance, for providing new strategies for the clinical treatment of stroke.
Collapse
Affiliation(s)
- Xiaoyun Xie
- Guangxi Medical University, The First Affiliated Hospital, Department of Neurology, Nanning, Guangxi, China.
| | - Jingli Liu
- Guangxi Medical University, The First Affiliated Hospital, Department of Neurology, Nanning, Guangxi, China.
| |
Collapse
|
8
|
Wang Y, Xia Y, Kou L, Yin S, Chi X, Li J, Sun Y, Wu J, Zhou Q, Zou W, Jin Z, Huang J, Xiong N, Wang T. Astrocyte-to-neuron reprogramming and crosstalk in the treatment of Parkinson's disease. Neurobiol Dis 2023:106224. [PMID: 37433411 DOI: 10.1016/j.nbd.2023.106224] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023] Open
Abstract
Parkinson's disease (PD) is currently the fastest growing disabling neurological disorder worldwide, with motor and non-motor symptoms being its main clinical manifestations. The primary pathological features include a reduction in the number of dopaminergic neurons in the substantia nigra and decrease in dopamine levels in the nigrostriatal pathway. Existing treatments only alleviate clinical symptoms and do not stop disease progression; slowing down the loss of dopaminergic neurons and stimulating their regeneration are emerging therapies. Preclinical studies have demonstrated that transplantation of dopamine cells generated from human embryonic or induced pluripotent stem cells can restore the loss of dopamine. However, the application of cell transplantation is limited owing to ethical controversies and the restricted source of cells. Until recently, the reprogramming of astrocytes to replenish lost dopaminergic neurons has provided a promising alternative therapy for PD. In addition, repair of mitochondrial perturbations, clearance of damaged mitochondria in astrocytes, and control of astrocyte inflammation may be extensively neuroprotective and beneficial against chronic neuroinflammation in PD. Therefore, this review primarily focuses on the progress and remaining issues in astrocyte reprogramming using transcription factors (TFs) and miRNAs, as well as exploring possible new targets for treating PD by repairing astrocytic mitochondria and reducing astrocytic inflammation.
Collapse
Affiliation(s)
- Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiulu Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
9
|
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
|
10
|
Chen J, Huang L, Yang Y, Xu W, Qin Q, Qin R, Liang X, Lai X, Huang X, Xie M, Chen L. Somatic Cell Reprogramming for Nervous System Diseases: Techniques, Mechanisms, Potential Applications, and Challenges. Brain Sci 2023; 13:brainsci13030524. [PMID: 36979334 PMCID: PMC10046178 DOI: 10.3390/brainsci13030524] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Nervous system diseases present significant challenges to the neuroscience community due to ethical and practical constraints that limit access to appropriate research materials. Somatic cell reprogramming has been proposed as a novel way to obtain neurons. Various emerging techniques have been used to reprogram mature and differentiated cells into neurons. This review provides an overview of somatic cell reprogramming for neurological research and therapy, focusing on neural reprogramming and generating different neural cell types. We examine the mechanisms involved in reprogramming and the challenges that arise. We herein summarize cell reprogramming strategies to generate neurons, including transcription factors, small molecules, and microRNAs, with a focus on different types of cells.. While reprogramming somatic cells into neurons holds the potential for understanding neurological diseases and developing therapeutic applications, its limitations and risks must be carefully considered. Here, we highlight the potential benefits of somatic cell reprogramming for neurological disease research and therapy. This review contributes to the field by providing a comprehensive overview of the various techniques used to generate neurons by cellular reprogramming and discussing their potential applications.
Collapse
Affiliation(s)
- Jiafeng Chen
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Lijuan Huang
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yue Yang
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Wei Xu
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qingchun Qin
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Rongxing Qin
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiaojun Liang
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xinyu Lai
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Nanning 530021, China
| | - Xiaoying Huang
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Minshan Xie
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Li Chen
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Nanning 530021, China
| |
Collapse
|
11
|
Probing Interleukin-6 in Stroke Pathology and Neural Stem Cell Transplantation. Int J Mol Sci 2022; 23:ijms232415453. [PMID: 36555094 PMCID: PMC9779061 DOI: 10.3390/ijms232415453] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Stem cell transplantation is historically understood as a powerful preclinical therapeutic following stroke models. Current clinical strategies including clot busting/retrieval are limited by their time windows (tissue plasminogen activator: 3-4 h) and inevitable reperfusion injuries. However, 24+ h post-stroke, stem cells reduce infarction size, improve neurobehavioral performance, and reduce inflammatory agents including interleukins. Typically, interleukin-6 (IL-6) is regarded as proinflammatory, and thus, preclinical studies often discuss it as beneficial for neurological recuperation when stem cells reduce IL-6's expression. However, some studies have also demonstrated neurological benefit with upregulation of IL-6 or preconditioning of stem cells with IL-6. This review specifically focuses on stem cells and IL-6, and their occasionally disparate, occasionally synergistic roles in the setting of ischemic cerebrovascular insults.
Collapse
|
12
|
Azeez IA, Awogbindin IO, Olayinka JN, Folarin RO, Adamu AS, Ior LD, Shehu AM, Mukhtar AI, Ajeigbe OF, Emokpae AO, Usende IL, Babatunde BR, Yusha'u Y, Olateju OI, Kamoga R, Benson AIO, Oparaji KC, Owemidu IO, Iliyasu MO, Imam MI, Olopade JO. Neural stem cell research in Africa: current realities and future prospects. Biol Open 2022; 11:280534. [PMID: 36326097 PMCID: PMC9641530 DOI: 10.1242/bio.059574] [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] [Indexed: 11/06/2022] Open
Abstract
Neural stem cells (NSCs) are immature progenitor cells that are found in developing and adult brains that have the potential of dividing actively and renewing themselves, with a complex form of gene expression. The generation of new brain cells in adult individuals was initially considered impossible, however, the landmark discovery of human neural stem cells in the hippocampus has been followed by further discoveries in other discreet regions of the brain. Investigation into the current state in Africa of the research and use of NSCs shows relatively limited activities on the continent. Information on the African application of NSCs for modelling disease mechanisms, drug discovery, and therapeutics is still limited. The International Brain Research Organization (IBRO)-African Regional Committee (ARC), with support from the Company of Biologists, and the Movement Disorder Society, sponsored the first African Basic School on NSC in Ibadan, Nigeria, with the vision of bringing together young neuroscientists and physicians across different fields in neuroscience to learn from leaders who have applied NSCs in stem cell research, the pathophysiology of neurodegenerative diseases, neuroanatomy, and neurotherapeutics. Twenty early-career researchers in academic institutions at junior and senior faculty cadres were selected from South Africa, Uganda and Nigeria. The students and organizer of the school, who wrote this review on the state of NSCs research in Africa, recommended the following: (1) other African countries can take a cue from South Africa and Nigeria in probing the phenomena of adult neurogenesis in unique animal species on the continent; (2) Africa should leverage the expertise and facilities of South African scientists and international collaborators in scaling up NSC research into these unique species and (3) Centers of Excellence should be established on the continent to serve as research hubs for training postgraduate students, and facilities for African scientists who trained overseas on NSCs.
Collapse
Affiliation(s)
- Idris A. Azeez
- Department of Veterinary Anatomy, University of Jos 1 , Jos, 930001 Nigeria
| | | | - Juliet N. Olayinka
- Department of Pharmacology and Therapeutics, Afe Babalola University 3 , Ado-Ekiti, 360001 Nigeria
| | - Royhaan O. Folarin
- Department of Anatomy, Olabisi Onabanjo University 4 , Ago-Iwoye, 120107 Nigeria
| | - Abubakar S. Adamu
- Department of Human Anatomy, Ahmadu Bello University 5 , Zaria, 810107 , Nigeria
| | - Lydia D. Ior
- Department of Pharmacology, University of Jos 6 , Jos, 930001 , Nigeria
| | - Asmau M. Shehu
- Department of Human Anatomy, Federal University Dutse 7 , Dutse, 720223 , Nigeria
- School of Anatomical Sciences, University of the Witwatersrand 8 , Johannesburg, Wits 2050 , South Africa
| | - Abubakar I. Mukhtar
- Department of Human Anatomy, Ahmadu Bello University 5 , Zaria, 810107 , Nigeria
| | - Olufunke F. Ajeigbe
- Elizade University, Ilara-Mokin, 340112 9 Department of Physical and Chemical Sciences, Biochemistry Programme , , Nigeria
| | | | - Ifukibot L. Usende
- Department of Veterinary Anatomy, University of Abuja 11 , Abuja, 900105 , Nigeria
| | | | - Yusuf Yusha'u
- Department of Human Physiology, Ahmadu Bello University 12 , Zaria, 810107 , Nigeria
| | - Oladiran I. Olateju
- School of Anatomical Sciences, University of the Witwatersrand 8 , Johannesburg, Wits 2050 , South Africa
| | - Ronald Kamoga
- Department of Pharmacology and Therapeutics, Mbarara University of Science and Technology 13 , Mbarara P.O. Box 1410 , Uganda
| | - Ayoola I. O. Benson
- Department of Human Anatomy, Elizade University, Ilara-Mokin 14 , Abakaliki, 482131 Nigeria
| | - Kenneth C. Oparaji
- Department of Physiology, Alex Ekwueme Federal University Ndufu-Alike 15 , Abakaliki, 482131 , Nigeria
| | - Idowu O. Owemidu
- Department of Physiology, Kogi State University 16 , Anyigba, 272102 , Nigeria
| | - Musa O. Iliyasu
- Department of Anatomy, Kogi State University 17 , Anyigba, 272102 , Nigeria
| | - Maryam I. Imam
- Department of Human Physiology, Ahmadu Bello University 12 , Zaria, 810107 , Nigeria
| | - James O. Olopade
- Department of Veterinary Anatomy, University of Ibadan 18 , Ibadan, 200005 , Nigeria
| |
Collapse
|
13
|
Guo W, Zhang X, Zhai J, Xue J. The roles and applications of neural stem cells in spinal cord injury repair. Front Bioeng Biotechnol 2022; 10:966866. [PMID: 36105599 PMCID: PMC9465243 DOI: 10.3389/fbioe.2022.966866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/28/2022] [Indexed: 12/05/2022] Open
Abstract
Spinal cord injury (SCI), which has no current cure, places a severe burden on patients. Stem cell-based therapies are considered promising in attempts to repair injured spinal cords; such options include neural stem cells (NSCs). NSCs are multipotent stem cells that differentiate into neuronal and neuroglial lineages. This feature makes NSCs suitable candidates for regenerating injured spinal cords. Many studies have revealed the therapeutic potential of NSCs. In this review, we discuss from an integrated view how NSCs can help SCI repair. We will discuss the sources and therapeutic potential of NSCs, as well as representative pre-clinical studies and clinical trials of NSC-based therapies for SCI repair.
Collapse
Affiliation(s)
- Wen Guo
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xindan Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Jiliang Zhai
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| |
Collapse
|
14
|
Aleksandrova MA, Sukhinich KK. Astrocytes of the Brain: Retinue Plays the King. Russ J Dev Biol 2022. [DOI: 10.1134/s1062360422040026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
15
|
Zhang SY, Zhao J, Ni JJ, Li H, Quan ZZ, Qing H. Application and prospects of high-throughput screening for in vitro neurogenesis. World J Stem Cells 2022; 14:393-419. [PMID: 35949394 PMCID: PMC9244953 DOI: 10.4252/wjsc.v14.i6.393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, high-throughput screening (HTS) has made great contributions to new drug discovery. HTS technology is equipped with higher throughput, minimized platforms, more automated and computerized operating systems, more efficient and sensitive detection devices, and rapid data processing systems. At the same time, in vitro neurogenesis is gradually becoming important in establishing models to investigate the mechanisms of neural disease or developmental processes. However, challenges remain in generating more mature and functional neurons with specific subtypes and in establishing robust and standardized three-dimensional (3D) in vitro models with neural cells cultured in 3D matrices or organoids representing specific brain regions. Here, we review the applications of HTS technologies on in vitro neurogenesis, especially aiming at identifying the essential genes, chemical small molecules and adaptive microenvironments that hold great prospects for generating functional neurons or more reproductive and homogeneous 3D organoids. We also discuss the developmental tendency of HTS technology, e.g., so-called next-generation screening, which utilizes 3D organoid-based screening combined with microfluidic devices to narrow the gap between in vitro models and in vivo situations both physiologically and pathologically.
Collapse
Affiliation(s)
- Shu-Yuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Jun-Jun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
16
|
Peng Z, Lu H, Yang Q, Xie Q. Astrocyte Reprogramming in Stroke: Opportunities and Challenges. Front Aging Neurosci 2022; 14:885707. [PMID: 35663583 PMCID: PMC9160982 DOI: 10.3389/fnagi.2022.885707] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022] Open
Abstract
Stroke is a major cause of morbidity and mortality worldwide. In the early stages of stroke, irreversible damage to neurons leads to high mortality and disability rates in patients. However, there are still no effective prevention and treatment measures for the resulting massive neuronal death in clinical practice. Astrocyte reprogramming has recently attracted much attention as an avenue for increasing neurons in mice after cerebral ischemia. However, the field of astrocyte reprogramming has recently been mired in controversy due to reports questioning whether newborn neurons are derived from astrocyte transformation. To better understand the process and controversies of astrocyte reprogramming, this review introduces the method of astrocyte reprogramming and its application in stroke. By targeting key transcription factors or microRNAs, astrocytes in the mouse brain could be reprogrammed into functional neurons. Additionally, we summarize some of the current controversies over the lack of cell lineage tracing and single-cell sequencing experiments to provide evidence of gene expression profile changes throughout the process of astrocyte reprogramming. Finally, we present recent advances in cell lineage tracing and single-cell sequencing, suggesting that it is possible to characterize the entire process of astrocyte reprogramming by combining these techniques.
Collapse
Affiliation(s)
- Zhouzhou Peng
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Hui Lu
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Qingwu Yang
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Qi Xie
- Department of Neurology, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
- *Correspondence: Qi Xie,
| |
Collapse
|
17
|
Generation of a Pure Culture of Neuron-like Cells with a Glutamatergic Phenotype from Mouse Astrocytes. Biomedicines 2022; 10:biomedicines10040928. [PMID: 35453678 PMCID: PMC9031297 DOI: 10.3390/biomedicines10040928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 12/04/2022] Open
Abstract
Astrocyte-to-neuron reprogramming is a promising therapeutic approach for treatment of neurodegenerative diseases. The use of small molecules as an alternative to the virus-mediated ectopic expression of lineage-specific transcription factors negates the tumorigenic risk associated with viral genetic manipulation and uncontrolled differentiation of stem cells. However, because previously developed methods for small-molecule reprogramming of astrocytes to neurons are multistep, complex, and lengthy, their applications in biomedicine, including clinical treatment, are limited. Therefore, our objective in this study was to develop a novel chemical-based approach to the cellular reprogramming of astrocytes into neurons with high efficiency and low complexity. To accomplish that, we used C8-D1a, a mouse astrocyte cell line, to assess the role of small molecules in reprogramming protocols that otherwise suffer from inconsistencies caused by variations in donor of the primary cell. We developed a new protocol by which a chemical mixture formulated with Y26732, DAPT, RepSox, CHIR99021, ruxolitinib, and SAG rapidly and efficiently induced the neural reprogramming of astrocytes in four days, with a conversion efficiency of 82 ± 6%. Upon exposure to the maturation medium, those reprogrammed cells acquired a glutaminergic phenotype over the next eleven days. We also demonstrated the neuronal functionality of the induced cells by confirming KCL-induced calcium flux.
Collapse
|
18
|
Yu G, Zhang Y, Ning B. Reactive Astrocytes in Central Nervous System Injury: Subgroup and Potential Therapy. Front Cell Neurosci 2022; 15:792764. [PMID: 35002629 PMCID: PMC8733560 DOI: 10.3389/fncel.2021.792764] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Traumatic central nervous system (CNS) injury, which includes both traumatic brain injury (TBI) and spinal cord injury (SCI), is associated with irreversible loss of neurological function and high medical care costs. Currently, no effective treatment exists to improve the prognosis of patients. Astrocytes comprise the largest population of glial cells in the CNS and, with the advancements in the field of neurology, are increasingly recognized as having key functions in both the brain and the spinal cord. When stimulated by disease or injury, astrocytes become activated and undergo a series of changes, including alterations in gene expression, hypertrophy, the loss of inherent functions, and the acquisition of new ones. Studies have shown that astrocytes are highly heterogeneous with respect to their gene expression profiles, and this heterogeneity accounts for their observed context-dependent phenotypic diversity. In the inured CNS, activated astrocytes play a dual role both as regulators of neuroinflammation and in scar formation. Identifying the subpopulations of reactive astrocytes that exert beneficial or harmful effects will aid in deciphering the pathological mechanisms underlying CNS injuries and ultimately provide a theoretical basis for the development of effective strategies for the treatment of associated conditions. Following CNS injury, as the disease progresses, astrocyte phenotypes undergo continuous changes. Although current research methods do not allow a comprehensive and accurate classification of astrocyte subpopulations in complex pathological contexts, they can nonetheless aid in understanding the roles of astrocytes in disease. In this review, after a brief introduction to the pathology of CNS injury, we summarize current knowledge regarding astrocyte activation following CNS injury, including: (a) the regulatory factors involved in this process; (b) the functions of different astrocyte subgroups based on the existing classification of astrocytes; and (c) attempts at astrocyte-targeted therapy.
Collapse
Affiliation(s)
- GuiLian Yu
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Zhang
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Ning
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
19
|
Ding Z, Dai C, Shan W, Liu R, Lu W, Gao W, Zhang H, Huang W, Guan J, Yin Z. TNF-α up-regulates Nanog by activating NF-κB pathway to induce primary rat spinal cord astrocytes dedifferentiation. Life Sci 2021; 287:120126. [PMID: 34758295 DOI: 10.1016/j.lfs.2021.120126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 10/10/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
AIMS Astrocytes re-acquire stem cell potential upon inflammation, thereby becoming a promising source of cells for regenerative medicine. Nanog is an essential transcription factor to maintain the characteristics of stem cells. We aimed to investigate the role of Nanog in astrocyte dedifferentiation. MAIN METHODS TNF-α was used to induce the dedifferentiation of primary rat spinal cord astrocytes. The expression of immature markers CD44 and Musashi-1 was detected by qRT-PCR and immunofluorescence. The Nanog gene is knocked down by small interference RNA. Nanog expression was measured by qRT-PCR and western blotting. BAY 11-7082 was used to suppress NF-κB signals in astrocytes. NF-κB signaling was evaluated by Western blotting. KEY FINDINGS Our results showed that TNF-α promoted the re-expression of CD44 and Musashi-1 in astrocytes. Dedifferentiated astrocytes could be induced to differentiate into oligodendrocyte lineage cells indicating that the astrocytes had pluripotency. In addition, TNF-α treatment activated NF-κB signaling pathway and up-regulated Nanog. Knockdown of Nanog reversed the increase of CD44 and Musashi-1 induced by TNF-α without affecting the activation of NF-κB signaling. Importantly, blocking NF-κB signaling by BAY 11-7082 inhibited the expression of immature markers suggesting that TNF-α induces dedifferentiation of astrocytes through the NF-κB signaling pathway. BAY 11-7082 could also inhibit the expression of Nanog, which indicated that Nanog was regulated by NF-κB signaling pathway. SIGNIFICANCE These findings indicate that activation of the NF-κB signaling pathway through TNF-α leads to astrocytes dedifferentiation via Nanog. These results expand our understanding of the mechanism of astrocytes dedifferentiation.
Collapse
Affiliation(s)
- Zhenfei Ding
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, 287#Chang Huai Road, Bengbu 230071, Anhui, China; Department of Orthopaedics, The Second People's Hospital of Hefei, Intersection of Guangde Road and Leshui Road, Hefei 230011, Anhui, China
| | - Ce Dai
- Department of Orthopaedics, The Second People's Hospital of Hefei, Intersection of Guangde Road and Leshui Road, Hefei 230011, Anhui, China
| | - Wenshan Shan
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Rui Liu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Wei Lu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Weilu Gao
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Hui Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Jianzhong Guan
- Department of Orthopaedics, The First Affiliated Hospital of Bengbu Medical College, 287#Chang Huai Road, Bengbu 230071, Anhui, China; Anhui Key Laboratory of Tissue Transplantation, 2600#Dong Hai Avenue, Bengbu 233030, Anhui, China.
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, 218#Ji Xi Road, Hefei 230032, Anhui, China.
| |
Collapse
|
20
|
Zhou C, Hu S, Botchway BOA, Zhang Y, Liu X. Valproic Acid: A Potential Therapeutic for Spinal Cord Injury. Cell Mol Neurobiol 2021; 41:1441-1452. [PMID: 32725456 PMCID: PMC11448682 DOI: 10.1007/s10571-020-00929-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
The lack of an effective pharmaceutical agent for spinal cord injury (SCI) is a current problematic situation for clinicians, as the rate of motor vehicle accidents among young adults is on the rise. SCI contributes to the high disability rate. Presently, evidences detailing the precise pathological mechanisms in SCI are limited, compounding to the unavailability of an effective treatment method. Surgery, though not a complete curative method, is useful in managing some of the associated symptoms of secondary SCI. Autophagy and inflammation are contributive factors to both exacerbation and improvement of SCI. The mammalian target of rapamycin (mTOR) signaling pathway is a key player in the regulation of inflammatory response and autophagy. Valproic acid (VPA), a clinically used antiepileptic drug, has been suggested to improve neurological conditions, including SCI. This report reviewed the correlation between mTOR and autophagy, as well as autophagy's role and the therapeutic effects of VPA in SCI. VPA regulates autophagy by potentially inhibiting mTORC1, a complex of mTOR, while also hindering inflammatory response. Conclusively, an effective treatment for SCI could lie in the timely regulation of mTOR signaling pathway, and VPA could be the potential drug that improves SCI owing to its propensity to regulate the mTOR signaling pathway.
Collapse
Affiliation(s)
- Conghui Zhou
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Songfeng Hu
- Department of Orthopedics, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing, 312000, Zhejiang Province, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| |
Collapse
|
21
|
Samoilova EM, Belopasov VV, Baklaushev VP. Transcription Factors of Direct Neuronal Reprogramming in Ontogenesis and Ex Vivo. Mol Biol 2021; 55:645-669. [DOI: 10.1134/s0026893321040087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 03/07/2025]
|
22
|
Wang Y, Zhang X, Chen F, Song N, Xie J. In vivo Direct Conversion of Astrocytes to Neurons Maybe a Potential Alternative Strategy for Neurodegenerative Diseases. Front Aging Neurosci 2021; 13:689276. [PMID: 34408642 PMCID: PMC8366583 DOI: 10.3389/fnagi.2021.689276] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/08/2021] [Indexed: 01/06/2023] Open
Abstract
Partly because of extensions in lifespan, the incidence of neurodegenerative diseases is increasing, while there is no effective approach to slow or prevent neuronal degeneration. As we all know, neurons cannot self-regenerate and may not be replaced once being damaged or degenerated in human brain. Astrocytes are widely distributed in the central nervous system (CNS) and proliferate once CNS injury or neurodegeneration occur. Actually, direct reprogramming astrocytes into functional neurons has been attracting more and more attention in recent years. Human astrocytes can be successfully converted into neurons in vitro. Notably, in vivo direct reprogramming of astrocytes into functional neurons were achieved in the adult mouse and non-human primate brains. In this review, we briefly summarized in vivo direct reprogramming of astrocytes into functional neurons as regenerative strategies for CNS diseases, mainly focusing on neurodegenerative diseases such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and Huntington’s disease (HD). We highlight and outline the advantages and challenges of direct neuronal reprogramming from astrocytes in vivo for future neuroregenerative medicine.
Collapse
Affiliation(s)
- Youcui Wang
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Xiaoqin Zhang
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Fenghua Chen
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Ning Song
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| | - Junxia Xie
- Institute of Brain Science and Disease, School of Basic Medicine, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
23
|
Hart CG, Karimi-Abdolrezaee S. Recent insights on astrocyte mechanisms in CNS homeostasis, pathology, and repair. J Neurosci Res 2021; 99:2427-2462. [PMID: 34259342 DOI: 10.1002/jnr.24922] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022]
Abstract
Astrocytes play essential roles in development, homeostasis, injury, and repair of the central nervous system (CNS). Their development is tightly regulated by distinct spatial and temporal cues during embryogenesis and into adulthood throughout the CNS. Astrocytes have several important responsibilities such as regulating blood flow and permeability of the blood-CNS barrier, glucose metabolism and storage, synapse formation and function, and axon myelination. In CNS pathologies, astrocytes also play critical parts in both injury and repair mechanisms. Upon injury, they undergo a robust phenotypic shift known as "reactive astrogliosis," which results in both constructive and deleterious outcomes. Astrocyte activation and migration at the site of injury provides an early defense mechanism to minimize the extent of injury by enveloping the lesion area. However, astrogliosis also contributes to the inhibitory microenvironment of CNS injury and potentiate secondary injury mechanisms, such as inflammation, oxidative stress, and glutamate excitotoxicity, which facilitate neurodegeneration in CNS pathologies. Intriguingly, reactive astrocytes are increasingly a focus in current therapeutic strategies as their activation can be modulated toward a neuroprotective and reparative phenotype. This review will discuss recent advancements in knowledge regarding the development and role of astrocytes in the healthy and pathological CNS. We will also review how astrocytes have been genetically modified to optimize their reparative potential after injury, and how they may be transdifferentiated into neurons and oligodendrocytes to promote repair after CNS injury and neurodegeneration.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
24
|
Vasan L, Park E, David LA, Fleming T, Schuurmans C. Direct Neuronal Reprogramming: Bridging the Gap Between Basic Science and Clinical Application. Front Cell Dev Biol 2021; 9:681087. [PMID: 34291049 PMCID: PMC8287587 DOI: 10.3389/fcell.2021.681087] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022] Open
Abstract
Direct neuronal reprogramming is an innovative new technology that involves the conversion of somatic cells to induced neurons (iNs) without passing through a pluripotent state. The capacity to make new neurons in the brain, which previously was not achievable, has created great excitement in the field as it has opened the door for the potential treatment of incurable neurodegenerative diseases and brain injuries such as stroke. These neurological disorders are associated with frank neuronal loss, and as new neurons are not made in most of the adult brain, treatment options are limited. Developmental biologists have paved the way for the field of direct neuronal reprogramming by identifying both intrinsic cues, primarily transcription factors (TFs) and miRNAs, and extrinsic cues, including growth factors and other signaling molecules, that induce neurogenesis and specify neuronal subtype identities in the embryonic brain. The striking observation that postmitotic, terminally differentiated somatic cells can be converted to iNs by mis-expression of TFs or miRNAs involved in neural lineage development, and/or by exposure to growth factors or small molecule cocktails that recapitulate the signaling environment of the developing brain, has opened the door to the rapid expansion of new neuronal reprogramming methodologies. Furthermore, the more recent applications of neuronal lineage conversion strategies that target resident glial cells in situ has expanded the clinical potential of direct neuronal reprogramming techniques. Herein, we present an overview of the history, accomplishments, and therapeutic potential of direct neuronal reprogramming as revealed over the last two decades.
Collapse
Affiliation(s)
- Lakshmy Vasan
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Eunjee Park
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Luke Ajay David
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Taylor Fleming
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
25
|
Wei ZYD, Shetty AK. Treating Parkinson's disease by astrocyte reprogramming: Progress and challenges. SCIENCE ADVANCES 2021; 7:7/26/eabg3198. [PMID: 34162545 PMCID: PMC8221613 DOI: 10.1126/sciadv.abg3198] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 05/07/2021] [Indexed: 05/04/2023]
Abstract
Parkinson's disease (PD), the second most prevalent neurodegenerative disorder, is typified by both motor and nonmotor symptoms. The current medications provide symptomatic relief but do not stimulate the production of new dopaminergic neurons in the substantia nigra. Astrocyte reprogramming has recently received much attention as an avenue for increasing functional dopaminergic neurons in the mouse PD brain. By targeting a microRNA (miRNA) loop, astrocytes in the mouse brain could be reprogrammed into functional dopaminergic neurons. Such in vivo astrocyte reprogramming in the mouse model of PD has successfully added new dopaminergic neurons to the substantia nigra and increased dopamine levels associated with axonal projections into the striatum. This review deliberates the astrocyte reprogramming methods using specific transcription factors and mRNAs and the progress in generating dopaminergic neurons in vivo. In addition, the translational potential, challenges, and potential risks of astrocyte reprogramming for an enduring alleviation of parkinsonian symptoms are conferred.
Collapse
Affiliation(s)
- Zhuang-Yao D Wei
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, Texas A&M University Health Science Center College of Medicine, College Station, TX, USA.
| |
Collapse
|
26
|
Zhang X, Chen F, Wang Y. Commentary: In vivo Neuroregeneration to Treat Ischemic Stroke Through NeuroD1 AAV-Based Gene Therapy in Adult Non-human Primates. Front Cell Dev Biol 2021; 9:648020. [PMID: 34124038 PMCID: PMC8194073 DOI: 10.3389/fcell.2021.648020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/12/2021] [Indexed: 01/21/2023] Open
Affiliation(s)
- Xiaoqin Zhang
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Fenghua Chen
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Youcui Wang
- Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Chiareli RA, Carvalho GA, Marques BL, Mota LS, Oliveira-Lima OC, Gomes RM, Birbrair A, Gomez RS, Simão F, Klempin F, Leist M, Pinto MCX. The Role of Astrocytes in the Neurorepair Process. Front Cell Dev Biol 2021; 9:665795. [PMID: 34113618 PMCID: PMC8186445 DOI: 10.3389/fcell.2021.665795] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are highly specialized glial cells responsible for trophic and metabolic support of neurons. They are associated to ionic homeostasis, the regulation of cerebral blood flow and metabolism, the modulation of synaptic activity by capturing and recycle of neurotransmitters and maintenance of the blood-brain barrier. During injuries and infections, astrocytes act in cerebral defense through heterogeneous and progressive changes in their gene expression, morphology, proliferative capacity, and function, which is known as reactive astrocytes. Thus, reactive astrocytes release several signaling molecules that modulates and contributes to the defense against injuries and infection in the central nervous system. Therefore, deciphering the complex signaling pathways of reactive astrocytes after brain damage can contribute to the neuroinflammation control and reveal new molecular targets to stimulate neurorepair process. In this review, we present the current knowledge about the role of astrocytes in brain damage and repair, highlighting the cellular and molecular bases involved in synaptogenesis and neurogenesis. In addition, we present new approaches to modulate the astrocytic activity and potentiates the neurorepair process after brain damage.
Collapse
Affiliation(s)
| | | | | | - Lennia Soares Mota
- Department of Pharmacology, Federal University of Goias, Goiânia, Brazil
| | | | | | - Alexander Birbrair
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renato Santiago Gomez
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Simão
- Research Division, Vascular Cell Biology, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | | | - Marcel Leist
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
28
|
Si Z, Wang X. Stem Cell Therapies in Alzheimer's Disease: Applications for Disease Modeling. J Pharmacol Exp Ther 2021; 377:207-217. [PMID: 33558427 DOI: 10.1124/jpet.120.000324] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/03/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with complex pathologic and biologic characteristics. Extracellular β-amyloid deposits, such as senile plaques, and intracellular aggregation of hyperphosphorylated tau, such as neurofibrillary tangles, remain the main neuropathological criteria for the diagnosis of AD. There is currently no effective treatment of the disease, and many clinical trials have failed to prove any benefits of new therapeutics. More recently, there has been increasing interest in harnessing the potential of stem cell technologies for drug discovery, disease modeling, and cell therapies, which have been used to study an array of human conditions, including AD. The recently developed and optimized induced pluripotent stem cell (iPSC) technology is a critical platform for screening anti-AD drugs and understanding mutations that modify AD. Neural stem cell (NSC) transplantation has been investigated as a new therapeutic approach to treat neurodegenerative diseases. Mesenchymal stem cells (MSCs) also exhibit considerable potential to treat neurodegenerative diseases by secreting growth factors and exosomes, attenuating neuroinflammation. This review highlights recent progress in stem cell research and the translational applications and challenges of iPSCs, NSCs, and MSCs as treatment strategies for AD. Even though these treatments are still in relative infancy, these developing stem cell technologies hold considerable promise to combat AD and other neurodegenerative disorders. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a neurodegenerative disease that results in learning and memory defects. Although some drugs have been approved for AD treatment, fewer than 20% of patients with AD benefit from these drugs. Therapies based on stem cells, including induced pluripotent stem cells, neural stem cells, and mesenchymal stem cells, provide promising therapeutic strategies for AD.
Collapse
Affiliation(s)
- Zizhen Si
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| | - Xidi Wang
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| |
Collapse
|
29
|
Mollinari C, Merlo D. Direct Reprogramming of Somatic Cells to Neurons: Pros and Cons of Chemical Approach. Neurochem Res 2021; 46:1330-1336. [PMID: 33666839 PMCID: PMC8084785 DOI: 10.1007/s11064-021-03282-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/31/2021] [Accepted: 02/20/2021] [Indexed: 12/11/2022]
Abstract
Translating successful preclinical research in neurodegenerative diseases into clinical practice has been difficult. The preclinical disease models used for testing new drugs not always appear predictive of the effects of the agents in the human disease state. Human induced pluripotent stem cells, obtained by reprogramming of adult somatic cells, represent a powerful system to study the molecular mechanisms of the disease onset and pathogenesis. However, these cells require a long time to differentiate into functional neural cells and the resetting of epigenetic information during reprogramming, might miss the information imparted by age. On the contrary, the direct conversion of somatic cells to neuronal cells is much faster and more efficient, it is safer for cell therapy and allows to preserve the signatures of donors’ age. Direct reprogramming can be induced by lineage-specific transcription factors or chemical cocktails and represents a powerful tool for modeling neurological diseases and for regenerative medicine. In this Commentary we present and discuss strength and weakness of several strategies for the direct cellular reprogramming from somatic cells to generate human brain cells which maintain age‐related features. In particular, we describe and discuss chemical strategy for cellular reprogramming as it represents a valuable tool for many applications such as aged brain modeling, drug screening and personalized medicine.
Collapse
Affiliation(s)
- Cristiana Mollinari
- Institute of Translational Pharmacology, National Research Council, Via Fosso del Cavaliere 100, 00133, Rome, Italy. .,Department of Neuroscience, Istituto Superiore di Sanita', Viale Regina Elena 299, 00161, Rome, Italy.
| | - Daniela Merlo
- Department of Neuroscience, Istituto Superiore di Sanita', Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
30
|
Rahman AA, Amruta N, Pinteaux E, Bix GJ. Neurogenesis After Stroke: A Therapeutic Perspective. Transl Stroke Res 2021; 12:1-14. [PMID: 32862401 PMCID: PMC7803692 DOI: 10.1007/s12975-020-00841-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Stroke is a major cause of death and disability worldwide. Yet therapeutic strategies available to treat stroke are very limited. There is an urgent need to develop novel therapeutics that can effectively facilitate functional recovery. The injury that results from stroke is known to induce neurogenesis in penumbra of the infarct region. There is considerable interest in harnessing this response for therapeutic purposes. This review summarizes what is currently known about stroke-induced neurogenesis and the factors that have been identified to regulate it. Additionally, some key studies in this field have been highlighted and their implications on future of stroke therapy have been discussed. There is a complex interplay between neuroinflammation and neurogenesis that dictates stroke outcome and possibly recovery. This highlights the need for a better understanding of the neuroinflammatory process and how it affects neurogenesis, as well as the need to identify new mechanisms and potential modulators. Neuroinflammatory processes and their impact on post-stroke repair have therefore also been discussed.
Collapse
Affiliation(s)
- Abir A Rahman
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Narayanappa Amruta
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, A.V. Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - Gregory J Bix
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
31
|
Niceforo A, Marioli C, Colasuonno F, Petrini S, Massey K, Tartaglia M, Bertini E, Moreno S, Compagnucci C. Altered cytoskeletal arrangement in induced pluripotent stem cells (iPSCs) and motor neurons from patients with riboflavin transporter deficiency. Dis Model Mech 2021; 14:dmm.046391. [PMID: 33468503 PMCID: PMC7927654 DOI: 10.1242/dmm.046391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/05/2021] [Indexed: 12/28/2022] Open
Abstract
The cytoskeletal network plays a crucial role in differentiation, morphogenesis, function and homeostasis of the nervous tissue, so that alterations in any of its components may lead to neurodegenerative diseases. Riboflavin transporter deficiency (RTD), a childhood-onset disorder characterized by degeneration of motor neurons (MNs), is caused by biallelic mutations in genes encoding the human riboflavin (RF) transporters. In a patient- specific induced Pluripotent Stem Cells (iPSCs) model of RTD, we recently demonstrated altered cell-cell contacts, energy dysmetabolism and redox imbalance.The present study focusses on cytoskeletal composition and dynamics associated to RTD, utilizing patients' iPSCs and derived MNs. Abnormal expression and distribution of α- and β-tubulin (α- and β-TUB), as well as imbalanced tyrosination of α-TUB, accompanied by impaired ability to repolymerize after nocodazole treatment, were found in RTD patient-derived iPSCs. Following differentiation, MNs showed consistent changes in TUB content, which was associated with abnormal morphofunctional features, such as neurite length and Ca++ homeostasis, suggesting impaired differentiation.Beneficial effects of RF supplementation, alone or in combination with the antioxidant molecule N-acetyl-cystine (NAC), were assessed. RF administration resulted in partially improved cytoskeletal features in patients' iPSCs and MNs, suggesting that redundancy of transporters may rescue cell functionality in the presence of adequate concentrations of the vitamin. Moreover, supplementation with NAC was demonstrated to be effective in restoring all the considered parameters, when used in combination with RF, thus supporting the therapeutic use of both compounds.
Collapse
Affiliation(s)
- Alessia Niceforo
- Department of Science, Laboratorio Interdipartimentale di Microscopia Elettronica, University Roma Tre, Rome 00146, Italy
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Chiara Marioli
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Fiorella Colasuonno
- Department of Science, Laboratorio Interdipartimentale di Microscopia Elettronica, University Roma Tre, Rome 00146, Italy
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Laboratories, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Keith Massey
- Science Director, Cure RTD Foundation, 6228 Northaven Road, Dallas, TX 75230, USA
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Enrico Bertini
- Department of Neuroscience, Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| | - Sandra Moreno
- Department of Science, Laboratorio Interdipartimentale di Microscopia Elettronica, University Roma Tre, Rome 00146, Italy
| | - Claudia Compagnucci
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy
| |
Collapse
|
32
|
He Y, An J, Yin JJ, Miao Q, Sui RX, Han QX, Ding ZB, Huang JJ, Ma CG, Xiao BG. Ethyl Pyruvate-Derived Transdifferentiation of Astrocytes to Oligodendrogenesis in Cuprizone-Induced Demyelinating Model. Neurotherapeutics 2021; 18:488-502. [PMID: 33140235 PMCID: PMC8116372 DOI: 10.1007/s13311-020-00947-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2020] [Indexed: 01/09/2023] Open
Abstract
Astrocytes redifferentiate into oligodendrogenesis, raising the possibility that astrocytes may be a potential target in the treatment of adult demyelinated lesion. Upon the basis of the improvement of behavior abnormality and demyelination by ethyl pyruvate (EP) treatment, we further explored whether EP affects the function of astrocytes, especially the transdifferentiation of astrocytes into oligodendrogenesis. The results showed that EP treatment increased the accumulation of astrocytes in myelin sheath and promoted the phagocytosis of myelin debris by astrocytes in vivo and in vitro. At the same time, EP treatment induced astrocytes to upregulate the expression of CNTF and BDNF in the corpus callosum and striatum as well as cultured astrocytes, accompanied by increased expression of nestin, Sox2, and β-catenin and decreased expression of Notch1 by astrocytes. As a result, EP treatment effectively promoted the generation of NG2+ and PDGF-Ra+ oligodendrocyte precursor cells (OPCs) that, in part, express astrocyte marker GFAP. Further confirmation was performed by intracerebral injection of primary astrocytes labeled with carboxyfluorescein diacetate succinimidyl ester (CFSE). As expected, NG2+ OPCs expressing CFSE and Sox2 were elevated in the corpus callosum of mice treated with EP following transplantation, revealing that EP can convert astrocytes into myelinating cells. Our results indicate the possibility that EP lead to effective myelin repair in patients suffering from myelination deficit.Graphical Abstract The diagram of EP action for promoting myelin regeneration in CPZ model. EP promoted migration and enrichment of astrocytes to demyelinated tissue and induced astrocytes to express neurotrophic CNTF and BDNF as well as translation factor nestin, Sox2, and β-catenin, which should contribute to astrocytes to differentiate of oligodendrogenesis. At the same time, EP promoted astrocytes to phagocytized myelin debris for removing the harmful substances of myelin regeneration.
Collapse
Affiliation(s)
- Yan He
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Jun An
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Jun-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Qiang Miao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Ruo-Xuan Sui
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Qing-Xian Han
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Zhi-Bin Ding
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Jian-Jun Huang
- Department of Neurology, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China.
- Department of Neurosurgery, First Hospital, Datong Coalmine Group, Datong, 037006, China.
| | - Bao-Guo Xiao
- Department of Neurology, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
33
|
Colasuonno F, Bertini E, Tartaglia M, Compagnucci C, Moreno S. Mitochondrial Abnormalities in Induced Pluripotent Stem Cells-Derived Motor Neurons from Patients with Riboflavin Transporter Deficiency. Antioxidants (Basel) 2020; 9:E1252. [PMID: 33317017 PMCID: PMC7763948 DOI: 10.3390/antiox9121252] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/31/2022] Open
Abstract
Riboflavin transporter deficiency (RTD) is a childhood-onset neurodegenerative disorder characterized by sensorineural deafness and motor neuron degeneration. Since riboflavin plays key functions in biological oxidation-reduction reactions, energy metabolism pathways involving flavoproteins are affected in RTD. We recently generated induced pluripotent stem cell (iPSC) lines from affected individuals as an in vitro model of the disease and documented mitochondrial impairment in these cells, dramatically impacting cell redox status. This work extends our study to motor neurons (MNs), i.e., the cell type most affected in patients with RTD. Altered intracellular distribution of mitochondria was detected by confocal microscopic analysis (following immunofluorescence for superoxide dismutase 2 (SOD2), as a dual mitochondrial and antioxidant marker), and βIII-Tubulin, as a neuronal marker. We demonstrate significantly lower SOD2 levels in RTD MNs, as compared to their healthy counterparts. Mitochondrial ultrastructural abnormalities were also assessed by focused ion beam/scanning electron microscopy. Moreover, we investigated the effects of combination treatment using riboflavin and N-acetylcysteine, which is a widely employed antioxidant. Overall, our findings further support the potential of patient-specific RTD models and provide evidence of mitochondrial alterations in RTD-related iPSC-derived MNs-emphasizing oxidative stress involvement in this rare disease. We also provide new clues for possible therapeutic strategies aimed at correcting mitochondrial defects, based on the use of antioxidants.
Collapse
Affiliation(s)
- Fiorella Colasuonno
- Department of Science, LIME, University of Roma Tre, 00146 Rome, Italy;
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Claudia Compagnucci
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Sandra Moreno
- Department of Science, LIME, University of Roma Tre, 00146 Rome, Italy;
| |
Collapse
|
34
|
Konstantinides N, Desplan C. Neuronal differentiation strategies: insights from single-cell sequencing and machine learning. Development 2020; 147:dev193631. [PMID: 33293292 PMCID: PMC7746664 DOI: 10.1242/dev.193631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neuronal replacement therapies rely on the in vitro differentiation of specific cell types from embryonic or induced pluripotent stem cells, or on the direct reprogramming of differentiated adult cells via the expression of transcription factors or signaling molecules. The factors used to induce differentiation or reprogramming are often identified by informed guesses based on differential gene expression or known roles for these factors during development. Moreover, differentiation protocols usually result in partly differentiated cells or the production of a mix of cell types. In this Hypothesis article, we suggest that, to overcome these inefficiencies and improve neuronal differentiation protocols, we need to take into account the developmental history of the desired cell types. Specifically, we present a strategy that uses single-cell sequencing techniques combined with machine learning as a principled method to select a sequence of programming factors that are important not only in adult neurons but also during differentiation.
Collapse
Affiliation(s)
| | - Claude Desplan
- Department of Biology, New York University, New York, NY 10003, USA
| |
Collapse
|
35
|
Lin Y, Anderson JD, Rahnama LMA, Gu SV, Knowlton AA. Exosomes in disease and regeneration: biological functions, diagnostics, and beneficial effects. Am J Physiol Heart Circ Physiol 2020; 319:H1162-H1180. [PMID: 32986962 PMCID: PMC7792703 DOI: 10.1152/ajpheart.00075.2020] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/30/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Exosomes are a subtype of extracellular vesicles. They range from 30 to 150 nm in diameter and originate from intraluminal vesicles. Exosomes were first identified as the mechanism for releasing unnecessary molecules from reticulocytes as they matured to red blood cells. Since then, exosomes have been shown to be secreted by a broad spectrum of cells and play an important role in the cardiovascular system. Different stimuli are associated with increased exosome release and result in different exosome content. The release of harmful DNA and other molecules via exosomes has been proposed as a mechanism to maintain cellular homeostasis. Because exosomes contain parent cell-specific proteins on the membrane and in the cargo that is delivered to recipient cells, exosomes are potential diagnostic biomarkers of various types of diseases, including cardiovascular disease. As exosomes are readily taken up by other cells, stem cell-derived exosomes have been recognized as a potential cell-free regenerative therapy to repair not only the injured heart but other tissues as well. The objective of this review is to provide an overview of the biological functions of exosomes in heart disease and tissue regeneration. Therefore, state-of-the-art methods for exosome isolation and characterization, as well as approaches to assess exosome functional properties, are reviewed. Investigation of exosomes provides a new approach to the study of disease and biological processes. Exosomes provide a potential "liquid biopsy," as they are present in most, if not all, biological fluids that are released by a wide range of cell types.
Collapse
Affiliation(s)
- Yun Lin
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | | | - Lily M A Rahnama
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | - Shenwen V Gu
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| | - Anne A Knowlton
- Molecular and Cellular Cardiology, Cardiovascular Medicine, University of California, Davis, California
| |
Collapse
|
36
|
Marioli C, Magliocca V, Petrini S, Niceforo A, Borghi R, Petrillo S, La Rosa P, Colasuonno F, Persichini T, Piemonte F, Massey K, Tartaglia M, Moreno S, Bertini E, Compagnucci C. Antioxidant Amelioration of Riboflavin Transporter Deficiency in Motoneurons Derived from Patient-Specific Induced Pluripotent Stem Cells. Int J Mol Sci 2020; 21:E7402. [PMID: 33036493 PMCID: PMC7582490 DOI: 10.3390/ijms21197402] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial dysfunction is a key element in the pathogenesis of neurodegenerative disorders, such as riboflavin transporter deficiency (RTD). This is a rare, childhood-onset disease characterized by motoneuron degeneration and caused by mutations in SLC52A2 and SLC52A3, encoding riboflavin (RF) transporters (RFVT2 and RFVT3, respectively), resulting in muscle weakness, ponto-bulbar paralysis and sensorineural deafness. Based on previous findings, which document the contribution of oxidative stress in RTD pathogenesis, we tested possible beneficial effects of several antioxidants (Vitamin C, Idebenone, Coenzyme Q10 and EPI-743, either alone or in combination with RF) on the morphology and function of neurons derived from induced pluripotent stem cells (iPSCs) from two RTD patients. To identify possible improvement of the neuronal morphotype, neurite length was measured by confocal microscopy after β-III tubulin immunofluorescent staining. Neuronal function was evaluated by determining superoxide anion generation by MitoSOX assay and intracellular calcium (Ca2+) levels, using the Fluo-4 probe. Among the antioxidants tested, EPI-743 restored the redox status, improved neurite length and ameliorated intracellular calcium influx into RTD motoneurons. In conclusion, we suggest that antioxidant supplementation may have a role in RTD treatment.
Collapse
Affiliation(s)
- Chiara Marioli
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (C.M.); (F.C.); (M.T.)
| | - Valentina Magliocca
- Department of Science, University Roma Tre, 00146 Rome, Italy; (V.M.); (T.P.)
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (A.N.); (R.B.); (S.P.); (F.P.); (E.B.)
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Research Laboratories, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Alessia Niceforo
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (A.N.); (R.B.); (S.P.); (F.P.); (E.B.)
- Department of Science, LIME, University Roma Tre, 00146 Rome, Italy;
| | - Rossella Borghi
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (A.N.); (R.B.); (S.P.); (F.P.); (E.B.)
- Department of Science, LIME, University Roma Tre, 00146 Rome, Italy;
| | - Sara Petrillo
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (A.N.); (R.B.); (S.P.); (F.P.); (E.B.)
| | - Piergiorgio La Rosa
- Department of Psychology, Division of Neuroscience, Sapienza University of Rome, 00185 Rome, Italy;
| | - Fiorella Colasuonno
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (C.M.); (F.C.); (M.T.)
- Department of Science, LIME, University Roma Tre, 00146 Rome, Italy;
| | - Tiziana Persichini
- Department of Science, University Roma Tre, 00146 Rome, Italy; (V.M.); (T.P.)
| | - Fiorella Piemonte
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (A.N.); (R.B.); (S.P.); (F.P.); (E.B.)
| | - Keith Massey
- Science Director, Cure RTD Foundation, 6228 Northaven Rd., Dallas, TX 75230, USA;
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (C.M.); (F.C.); (M.T.)
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, 00146 Rome, Italy;
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Department of Neuroscience, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (A.N.); (R.B.); (S.P.); (F.P.); (E.B.)
| | - Claudia Compagnucci
- Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy; (C.M.); (F.C.); (M.T.)
| |
Collapse
|
37
|
Morales-Garcia JA, Calleja-Conde J, Lopez-Moreno JA, Alonso-Gil S, Sanz-SanCristobal M, Riba J, Perez-Castillo A. N,N-dimethyltryptamine compound found in the hallucinogenic tea ayahuasca, regulates adult neurogenesis in vitro and in vivo. Transl Psychiatry 2020; 10:331. [PMID: 32989216 PMCID: PMC7522265 DOI: 10.1038/s41398-020-01011-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
N,N-dimethyltryptamine (DMT) is a component of the ayahuasca brew traditionally used for ritual and therapeutic purposes across several South American countries. Here, we have examined, in vitro and vivo, the potential neurogenic effect of DMT. Our results demonstrate that DMT administration activates the main adult neurogenic niche, the subgranular zone of the dentate gyrus of the hippocampus, promoting newly generated neurons in the granular zone. Moreover, these mice performed better, compared to control non-treated animals, in memory tests, which suggest a functional relevance for the DMT-induced new production of neurons in the hippocampus. Interestingly, the neurogenic effect of DMT appears to involve signaling via sigma-1 receptor (S1R) activation since S1R antagonist blocked the neurogenic effect. Taken together, our results demonstrate that DMT treatment activates the subgranular neurogenic niche regulating the proliferation of neural stem cells, the migration of neuroblasts, and promoting the generation of new neurons in the hippocampus, therefore enhancing adult neurogenesis and improving spatial learning and memory tasks.
Collapse
Affiliation(s)
- Jose A. Morales-Garcia
- grid.4711.30000 0001 2183 4846Institute for Biomedical Research “A. Sols” (CSIC-UAM). Arturo Duperier 4, 28029 Madrid, Spain ,grid.413448.e0000 0000 9314 1427Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031 Madrid, Spain ,grid.4795.f0000 0001 2157 7667Department of Cellular Biology, School of Medicine, Complutense University of Madrid, Plaza Ramón y Cajal, 28040 Madrid, Spain ,grid.411347.40000 0000 9248 5770Cellular Neurobiology Laboratory, Neurobiology Department, UCS-UCM, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Javier Calleja-Conde
- grid.4795.f0000 0001 2157 7667Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Carretera de Humera, 28223 Madrid, Spain
| | - Jose A. Lopez-Moreno
- grid.4795.f0000 0001 2157 7667Department of Psychobiology and Behavioural Sciences Methods, Faculty of Psychology, Complutense University of Madrid, Carretera de Humera, 28223 Madrid, Spain
| | - Sandra Alonso-Gil
- grid.4711.30000 0001 2183 4846Institute for Biomedical Research “A. Sols” (CSIC-UAM). Arturo Duperier 4, 28029 Madrid, Spain ,grid.413448.e0000 0000 9314 1427Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031 Madrid, Spain
| | - Marina Sanz-SanCristobal
- grid.4711.30000 0001 2183 4846Institute for Biomedical Research “A. Sols” (CSIC-UAM). Arturo Duperier 4, 28029 Madrid, Spain ,grid.413448.e0000 0000 9314 1427Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031 Madrid, Spain
| | - Jordi Riba
- grid.5012.60000 0001 0481 6099Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Ana Perez-Castillo
- Institute for Biomedical Research "A. Sols" (CSIC-UAM). Arturo Duperier 4, 28029, Madrid, Spain. .,Spanish Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), c/ Valderrebollo 5, 28031, Madrid, Spain. .,Cellular Neurobiology Laboratory, Neurobiology Department, UCS-UCM, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain.
| |
Collapse
|
38
|
Yao Y, Wang C. Dedifferentiation: inspiration for devising engineering strategies for regenerative medicine. NPJ Regen Med 2020; 5:14. [PMID: 32821434 PMCID: PMC7395755 DOI: 10.1038/s41536-020-00099-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
Cell dedifferentiation is the process by which cells grow reversely from a partially or terminally differentiated stage to a less differentiated stage within their own lineage. This extraordinary phenomenon, observed in many physiological processes, inspires the possibility of developing new therapeutic approaches to regenerate damaged tissue and organs. Meanwhile, studies also indicate that dedifferentiation can cause pathological changes. In this review, we compile the literature describing recent advances in research on dedifferentiation, with an emphasis on tissue-specific findings, cellular mechanisms, and potential therapeutic applications from an engineering perspective. A critical understanding of such knowledge may provide fresh insights for designing new therapeutic strategies for regenerative medicine based on the principle of cell dedifferentiation.
Collapse
Affiliation(s)
- Yongchang Yao
- Department of Joint Surgery, The First Affiliated Hospital of Guangzhou Medical University, 510120 Guangzhou, China.,Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Guangzhou, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
39
|
Nizzardo M, Taiana M, Rizzo F, Aguila Benitez J, Nijssen J, Allodi I, Melzi V, Bresolin N, Comi GP, Hedlund E, Corti S. Synaptotagmin 13 is neuroprotective across motor neuron diseases. Acta Neuropathol 2020; 139:837-853. [PMID: 32065260 PMCID: PMC7181443 DOI: 10.1007/s00401-020-02133-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/27/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022]
Abstract
In amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), spinal and lower brainstem motor neurons degenerate, but some motor neuron subtypes are spared, including oculomotor neurons (OMNs). The mechanisms responsible for this selective degeneration are largely unknown, but the molecular signatures of resistant and vulnerable motor neurons are distinct and offer clues to neuronal resilience and susceptibility. Here, we demonstrate that healthy OMNs preferentially express Synaptotagmin 13 (SYT13) compared to spinal motor neurons. In end-stage ALS patients, SYT13 is enriched in both OMNs and the remaining relatively resilient spinal motor neurons compared to controls. Overexpression of SYT13 in ALS and SMA patient motor neurons in vitro improves their survival and increases axon lengths. Gene therapy with Syt13 prolongs the lifespan of ALS mice by 14% and SMA mice by 50% by preserving motor neurons and delaying muscle denervation. SYT13 decreases endoplasmic reticulum stress and apoptosis of motor neurons, both in vitro and in vivo. Thus, SYT13 is a resilience factor that can protect motor neurons and a candidate therapeutic target across motor neuron diseases.
Collapse
|
40
|
Kwak TH, Hali S, Kim S, Kim J, La H, Kim KP, Hong KH, Shin CY, Kim NH, Han DW. Robust and Reproducible Generation of Induced Neural Stem Cells from Human Somatic Cells by Defined Factors. Int J Stem Cells 2020; 13:80-92. [PMID: 32114739 PMCID: PMC7119206 DOI: 10.15283/ijsc19097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Recent studies have described direct reprogramming of mouse and human somatic cells into induced neural stem cells (iNSCs) using various combinations of transcription factors. Although iNSC technology holds a great potential for clinical applications, the low conversion efficiency and limited reproducibility of iNSC generation hinder its further translation into the clinic, strongly suggesting the necessity of highly reproducible method for human iNSCs (hiNSCs). Thus, in orderto develop a highly efficient and reproducible protocol for hiNSC generation, we revisited the reprogramming potentials of previously reported hiNSC reprogramming cocktails by comparing the reprogramming efficiency of distinct factor combinations including ours. METHODS We introduced distinct factor combinations, OSKM (OCT4+SOX2+KLF4+C-MYC), OCT4 alone, SOX2 alone, SOX2+HMGA2, BRN4+SKM+SV40LT (BSKMLT), SKLT, SMLT, and SKMLT and performed comparative analysis of reprogramming potentials of distinct factor combinations in hiNSC generation. RESULTS Here we show that ectopic expression of five reprogramming factors, BSKMLT leads the robust hiNSC generation (>80 folds enhanced efficiency) from human somatic cells compared with previously described factor combinations. With our combination, we were able to observe hiNSC conversion within 7 days of transduction. Throughout further optimization steps, we found that both BRN4 and KLF4 are not essential for hiNSC conversion. CONCLUSIONS Our factor combination could robustly and reproducibly generate hiNSCs from human somatic cells with distinct origins. Therefore, our novel reprogramming strategy might serve as a useful tool for hiNSC-based clinical application.
Collapse
Affiliation(s)
- Tae Hwan Kwak
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, Korea
| | - Sai Hali
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, Korea
| | - Sungmin Kim
- School of Cell and Molecular Medicine, University of Bristol, Bristol, UK
| | - Jonghun Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Hyeonwoo La
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul, Korea
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kwon Ho Hong
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Seoul, Korea
| | - Chan Young Shin
- Department of Neuroscience, School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul, Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences, Chungbuk National University, Cheongju, Korea
| | - Dong Wook Han
- School of Biotechnology and Healthcare, Wuyi University, Jiangmen, China
| |
Collapse
|
41
|
Liu K, Ma W, Li C, Li J, Zhang X, Liu J, Liu W, Wu Z, Zang C, Liang Y, Guo J, Li L. Advances in Transcription Factors Related to Neuroglial Cell Reprogramming. Transl Neurosci 2020; 11:17-27. [PMID: 32161682 PMCID: PMC7053399 DOI: 10.1515/tnsci-2020-0004] [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: 08/23/2019] [Accepted: 01/07/2020] [Indexed: 11/27/2022] Open
Abstract
Neuroglial cells have a high level of plasticity, and many types of these cells are present in the nervous system. Neuroglial cells provide diverse therapeutic targets for neurological diseases and injury repair. Cell reprogramming technology provides an efficient pathway for cell transformation during neural regeneration, while transcription factor-mediated reprogramming can facilitate the understanding of how neuroglial cells mature into functional neurons and promote neurological function recovery.
Collapse
Affiliation(s)
- Kuangpin Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Chunyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Junjun Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Xingkui Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Jie Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Liu
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Zheng Wu
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Chenghao Zang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yu Liang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| | - Jianhui Guo
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Liyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
42
|
Xia X, Li C, Wang Y, Deng X, Ma Y, Ding L, Zheng J. Reprogrammed astrocytes display higher neurogenic competence, migration ability and cell death resistance than reprogrammed fibroblasts. Transl Neurodegener 2020; 9:6. [PMID: 32071715 PMCID: PMC7011554 DOI: 10.1186/s40035-020-0184-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 01/30/2020] [Indexed: 12/20/2022] Open
Abstract
The direct reprogramming of somatic cells into induced neural progenitor cells (iNPCs) has been envisioned as a promising approach to overcome ethical and clinical issues of pluripotent stem cell transplantation. We previously reported that astrocyte-derived induced pluripotent stem cells (iPSCs) have more tendencies for neuronal differentiation than fibroblast-derived iPSCs. However, the differences of neurogenic potential between astrocyte-derived iNPCs (AiNPCs) and iNPCs from non-neural origins, such as fibroblast-derived iNPCs (FiNPCs), and the underlying mechanisms remain unclear. Our results suggested that AiNPCs exhibited higher differentiation efficiency, mobility and survival capacities, compared to FiNPCs. The whole transcriptome analysis revealed higher activities of TGFβ signaling in AiNPCs, versus FiNPCs, following a similar trend between astrocytes and fibroblasts. The higher neurogenic competence, migration ability, and cell death resistance of AiNPCs could be abrogated using TGFβ signaling inhibitor LY2157299. Hence, our study demonstrates the difference between iNPCs generated from neural and non-neural cells, together with the underlying mechanisms, which, provides valuable information for donor cell selection in the reprogramming approach.
Collapse
Affiliation(s)
- Xiaohuan Xia
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Chunhong Li
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yi Wang
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xiaobei Deng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yizhao Ma
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Lu Ding
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Jialin Zheng
- 1Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China.,2Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092 China.,3Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA.,4Department of Pathology and Microbiology, University of Nebraska Medical Center,, Omaha, NE 68198-5930 USA
| |
Collapse
|
43
|
Reprogramming Fibroblasts to Neural Stem Cells by Overexpression of the Transcription Factor Ptf1a. Methods Mol Biol 2020; 2117:245-263. [PMID: 31960384 DOI: 10.1007/978-1-0716-0301-7_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Neural stem cells (NSCs) have the features of both neural progenitors and stem cells, and show great potentials in translational research and regenerative medicine. Studies on NSCs have been greatly accelerated by the introduction of induced neural stem cells (iNSCs). The iNSCs are usually differentiated from induced pluripotent stem cells (iPSCs) or transdifferentiated from somatic cells such as fibroblasts or glial cells. Here, we describe a detailed protocol to reprogram human and mouse fibroblasts into iNSCs by overexpression of a transcription factor Ptf1a delivered by lentiviruses. The obtained iNSC lines have a strong self-renewal ability and are capable of differentiating into various types of neurons, astrocytes, and oligodendrocytes both in vitro and in vivo. The protocol is quite simple but powerful to produce iNSC lines.
Collapse
|
44
|
Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer's disease with iPSC-derived brain cells. Mol Psychiatry 2020; 25:148-167. [PMID: 31391546 PMCID: PMC6906186 DOI: 10.1038/s41380-019-0468-3] [Citation(s) in RCA: 288] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/10/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is a devastating neurodegenerative disorder with no cure. Countless promising therapeutics have shown efficacy in rodent Alzheimer's disease models yet failed to benefit human patients. While hope remains that earlier intervention with existing therapeutics will improve outcomes, it is becoming increasingly clear that new approaches to understand and combat the pathophysiology of Alzheimer's disease are needed. Human induced pluripotent stem cell (iPSC) technologies have changed the face of preclinical research and iPSC-derived cell types are being utilized to study an array of human conditions, including neurodegenerative disease. All major brain cell types can now be differentiated from iPSCs, while increasingly complex co-culture systems are being developed to facilitate neuroscience research. Many cellular functions perturbed in Alzheimer's disease can be recapitulated using iPSC-derived cells in vitro, and co-culture platforms are beginning to yield insights into the complex interactions that occur between brain cell types during neurodegeneration. Further, iPSC-based systems and genome editing tools will be critical in understanding the roles of the numerous new genes and mutations found to modify Alzheimer's disease risk in the past decade. While still in their relative infancy, these developing iPSC-based technologies hold considerable promise to push forward efforts to combat Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jay Penney
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - William T Ralvenius
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
45
|
Abstract
The details of adult neurogenesis, including environmental triggers, region specificity, and species homology remain an area of intense investigation. Slowing or halting age-related cognitive dysfunction, or restoring neurons lost to disease or injury represent just a fraction of potential therapeutic applications. New neurons can derive from stem cells, pluripotent neural progenitor cells, or non-neuronal glial cells, such as astrocytes. Astrocytes must be epigenetically “reprogrammed” to become neurons, which can occur both naturally in vivo, and via artificial exogenous treatments. While neural progenitor cells are localized to a few neurogenic zones in the adult brain, astrocytes populate almost every brain structure. In this review, we will summarize recent research into neurogenesis that arises from conversion of post-mitotic astrocytes, detail the genetic and epigenetic pathways that regulate this process, and discuss the possible clinical relevance in supplementing stem-cell neurogenic therapies.
Collapse
Affiliation(s)
- Brian B Griffiths
- Department of Anesthesiology, Pain & Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Anvee Bhutani
- Department of Anesthesiology, Pain & Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Creed M Stary
- Department of Anesthesiology, Pain & Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
46
|
Aravantinou-Fatorou K, Thomaidou D. In Vitro Direct Reprogramming of Mouse and Human Astrocytes to Induced Neurons. Methods Mol Biol 2020; 2155:41-61. [PMID: 32474866 DOI: 10.1007/978-1-0716-0655-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Direct neuronal reprogramming, rewiring the epigenetic and transcriptional network of a differentiated cell type to neuron, apart from being a very promising approach for the treatment of brain injury and neurodegeneration, also offers a prime opportunity to investigate the molecular underpinnings of neuronal cell fate determination, as the precise molecular mechanisms that establish neuronal fate and diversity at the transcriptional and epigenetic level are incompletely understood. Recent studies from a number of groups, including ours, have shown that astrocytes can be directly reprogrammed into functional neurons in vitro and in vivo following ectopic overexpression of combinations of transcription factors, neurogenic proteins, miRNAs, and small chemical molecules.In this chapter we describe the protocols for in vitro converting primary cortical astrocytes of mouse and human origin to induced neurons, through forced expression of two neurogenic molecules, either each one alone or in combination: the master regulatory bHLH proneural transcription factor NEUROGENIN-2 (NEUROG2) and the neurogenic protein CEND1. Forced expression of each one of the two neurogenic proteins in primary astrocytes via retroviral gene transfer results in their direct conversion to subtype-specific induced neurons, while simultaneous coexpression of both molecules drives them predominantly toward acquisition of a neural precursor cell (NPC) state. Although mouse and human astrocytes exhibit differences in their reprogramming rate and particular characteristics, they can both get efficiently in vitro transdifferentiated to NPCs and induced neurons upon NEUROG2 or/and CEND1 forced expression using the reprogramming protocols described in the chapter, presenting valuable cellular platforms for mechanistic studies and in vivo applications to restore neurodegeneration.
Collapse
Affiliation(s)
- Katerina Aravantinou-Fatorou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Dimitra Thomaidou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece.
| |
Collapse
|
47
|
Gam R, Sung M, Prasad Pandurangan A. Experimental and Computational Approaches to Direct Cell Reprogramming: Recent Advancement and Future Challenges. Cells 2019; 8:E1189. [PMID: 31581647 PMCID: PMC6829265 DOI: 10.3390/cells8101189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/26/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
The process of direct cell reprogramming, also named transdifferentiation, permits for the conversion of one mature cell type directly into another, without returning to a dedifferentiated state. This makes direct reprogramming a promising approach for the development of several cellular and tissue engineering therapies. To achieve the change in the cell identity, direct reprogramming requires an arsenal of tools that combine experimental and computational techniques. In the recent years, several methods of transdifferentiation have been developed. In this review, we will introduce the concept of direct cell reprogramming and its background, and cover the recent developments in the experimental and computational prediction techniques with their applications. We also discuss the challenges of translating this technology to clinical setting, accompanied with potential solutions.
Collapse
Affiliation(s)
- Rihab Gam
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Minkyung Sung
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | | |
Collapse
|
48
|
Yavarpour‐Bali H, Nakhaei‐Nejad M, Yazdi A, Ghasemi‐Kasman M. Direct conversion of somatic cells towards oligodendroglial lineage cells: A novel strategy for enhancement of myelin repair. J Cell Physiol 2019; 235:2023-2036. [DOI: 10.1002/jcp.29195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | | | - Azadeh Yazdi
- Department of Physiology, Faculty of Medical Sciences Isfahan University of Medical Sciences, Isfahan Iran
| | - Maryam Ghasemi‐Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Neuroscience Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
| |
Collapse
|
49
|
Flitsch LJ, Brüstle O. Evolving principles underlying neural lineage conversion and their relevance for biomedical translation. F1000Res 2019; 8. [PMID: 31559012 PMCID: PMC6743253 DOI: 10.12688/f1000research.18926.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
Scientific and technological advances of the past decade have shed light on the mechanisms underlying cell fate acquisition, including its transcriptional and epigenetic regulation during embryonic development. This knowledge has enabled us to purposefully engineer cell fates
in vitro by manipulating expression levels of lineage-instructing transcription factors. Here, we review the state of the art in the cell programming field with a focus on the derivation of neural cells. We reflect on what we know about the mechanisms underlying fate changes in general and on the degree of epigenetic remodeling conveyed by the distinct reprogramming and direct conversion strategies available. Moreover, we discuss the implications of residual epigenetic memory for biomedical applications such as disease modeling and neuroregeneration. Finally, we cover recent developments approaching cell fate conversion in the living brain and define questions which need to be addressed before cell programming can become an integral part of translational medicine.
Collapse
Affiliation(s)
- Lea Jessica Flitsch
- Institute of Reconstructive Neurobiology, University of Bonn School of Medicine & University Hospital Bonn, Bonn, North Rhine Wesphalia, 53127, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn School of Medicine & University Hospital Bonn, Bonn, North Rhine Wesphalia, 53127, Germany
| |
Collapse
|
50
|
Yang H, Liu C, Fan H, Chen B, Huang D, Zhang L, Zhang Q, An J, Zhao J, Wang Y, Hao D. Sonic Hedgehog Effectively Improves Oct4-Mediated Reprogramming of Astrocytes into Neural Stem Cells. Mol Ther 2019; 27:1467-1482. [PMID: 31153826 PMCID: PMC6698197 DOI: 10.1016/j.ymthe.2019.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 01/19/2023] Open
Abstract
Irreversible neuron loss following spinal cord injury (SCI) usually results in persistent neurological dysfunction. The generation of autologous neural stem cells (NSCs) holds great potential for neural replenishment therapies and drug screening in SCI. Our recent studies demonstrated that mature astrocytes from the spinal cord can directly revert back to a pluripotent state under appropriate signals. However, in previous attempts, the reprogramming of astrocytes into induced NSCs (iNSCs) was unstable, inefficient, and frequently accompanied by generation of intermediate precursors. It remained unknown how to further increase the efficiency of astrocyte reprogramming into iNSCs. Here, we show that mature astrocytes could be directly converted into iNSCs by a single transcription factor, Oct4, and that the iNSCs displayed typical neurosphere morphology, authentic NSC gene expression, self-renewal capacity, and multipotency. Strikingly, Oct4-driven reprogramming of astrocytes into iNSCs was potentiated with continuous sonic hedgehog (Shh) stimulation, as demonstrated by a sped-up reprogramming and increased conversion efficiency. Moreover, the iNSC-derived neurons possessed functionality as neurons. Importantly, crosstalk between Sox2/Shh-targeted downstream signals and phosphatidylinositol 3-kinase/cyclin-dependent kinase 2/Smad ubiquitin regulatory factor 2 (PI3K/Cdk2/Smurf2) signaling is likely involved in the mechanisms underlying this cellular event. The highly efficient reprogramming of astrocytes to generate iNSCs will provide an alternative therapeutic approach for SCI using autologous cells.
Collapse
Affiliation(s)
- Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China.
| | - Cuicui Liu
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Hong Fan
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Bo Chen
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Dageng Huang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Lingling Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Qian Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Jing An
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Jingjing Zhao
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Yi Wang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Dingjun Hao
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China; Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China.
| |
Collapse
|