1
|
Panferov E, Dodina M, Reshetnikov V, Ryapolova A, Ivanov R, Karabelsky A, Minskaia E. Induced Pluripotent (iPSC) and Mesenchymal (MSC) Stem Cells for In Vitro Disease Modeling and Regenerative Medicine. Int J Mol Sci 2025; 26:5617. [PMID: 40565081 DOI: 10.3390/ijms26125617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 06/04/2025] [Accepted: 06/10/2025] [Indexed: 06/28/2025] Open
Abstract
In vitro disease modeling can be used both for understanding the development of pathology and for screening various therapies, such as gene therapies. This approach decreases costs, shortens research timelines, reduces animal testing, and may be more accurate in replicating the disease phenotype compared to animal models. This review focuses on the two types of stem cells: induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs), which can be used for this purpose. Special attention is given to the impact of the isolation source and the variable expression of certain phenotypic markers on the differentiation capacity of these cells. Both similarities and critical differences between iPSCs and MSCs, as well as the outcomes of past and ongoing clinical trials, are discussed in order to gain insight and understanding as to which of these two cell types can be more suitable for the particular biomedical application.
Collapse
Affiliation(s)
- Egor Panferov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| | - Maria Dodina
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| | - Vasiliy Reshetnikov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| | - Anastasia Ryapolova
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| | - Roman Ivanov
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| | - Alexander Karabelsky
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| | - Ekaterina Minskaia
- Translational Medicine Research Center, Sirius University of Science and Technology, Olympic Avenue, 1, 354340 Sochi, Russia
| |
Collapse
|
2
|
Zikmund T, Fiorentino J, Penfold C, Stock M, Shpudeiko P, Agarwal G, Langfeld L, Petrova K, Peshkin L, Hamperl S, Scialdone A, Hoermanseder E. Differentiation success of reprogrammed cells is heterogeneous in vivo and modulated by somatic cell identity memory. Stem Cell Reports 2025; 20:102447. [PMID: 40086446 PMCID: PMC12069884 DOI: 10.1016/j.stemcr.2025.102447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Nuclear reprogramming can change cellular fates. Yet, reprogramming efficiency is low, and the resulting cell types are often not functional. Here, we used nuclear transfer to eggs to follow single cells during reprogramming in vivo. We show that the differentiation success of reprogrammed cells varies across cell types and depends on the expression of genes specific to the previous cellular identity. We find subsets of reprogramming-resistant cells that fail to form functional cell types, undergo cell death, or disrupt normal body patterning. Reducing expression levels of genes specific to the cell type of origin leads to better reprogramming and improved differentiation trajectories. Thus, our work demonstrates that failing to reprogram in vivo is cell type specific and emphasizes the necessity of minimizing aberrant transcripts of the previous somatic identity for improving reprogramming.
Collapse
Affiliation(s)
- Tomas Zikmund
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany
| | - Jonathan Fiorentino
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Chris Penfold
- Wellcome Trust/Cancer Research, Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Marco Stock
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Polina Shpudeiko
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Gaurav Agarwal
- Wellcome Trust/Cancer Research, Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Larissa Langfeld
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany
| | - Kseniya Petrova
- Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Leonid Peshkin
- Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephan Hamperl
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany.
| | - Eva Hoermanseder
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany.
| |
Collapse
|
3
|
Morichon L, Assou S, Bourdin A, Muriaux D, De Vos J. [In vitro preclinical models reproducing the respiratory epithelium: Application to the study of SARS-CoV-2 virus infection]. Rev Mal Respir 2025; 42:153-158. [PMID: 40023713 DOI: 10.1016/j.rmr.2025.02.001] [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: 03/04/2025]
Abstract
Highlighted by the COVID-19 pandemic, the study of respiratory infections is a global health priority. To this end, many preclinical in vitro study models have been developed to reproduce nasal, bronchial or alveolar respiratory epithelium. These models can be established from immortalised cell lines, primary culture or induced pluripotent stem cells (iPSC). They can also be constructed in various three-dimensional structures that are more or less physiological and easy to use. This synthetic review puts into perspective the advantages and limitations of these models, while highlighting their relevance for the study of the mechanisms of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- L Morichon
- Centre d'étude des maladies infectieuses et pharmacologie anti-infectieuses (CEMIPAI), CNRS UAR3725, Montpellier, France; Institute of Regenerative Medecine and Biotherapy (IRMB), CHU de Montpellier, Inserm U1183, université de Montpellier, Montpellier, France.
| | - S Assou
- Institute of Regenerative Medecine and Biotherapy (IRMB), CHU de Montpellier, Inserm U1183, université de Montpellier, Montpellier, France
| | - A Bourdin
- Department of Respiratory Diseases, Inserm, Arnaud-de-Villeneuve Hospital, CHU de Montpellier, Montpellier, France; PhyMedExp, Inserm U1046, CNRS UMR 9214, University of Montpellier, Montpellier, France
| | - D Muriaux
- Centre d'étude des maladies infectieuses et pharmacologie anti-infectieuses (CEMIPAI), CNRS UAR3725, Montpellier, France; Institut de recherche en infectiologie à Montpellier (IRIM), UMR 9004, CNRS & université de Montpellier, Montpellier, France
| | - J De Vos
- Institute of Regenerative Medecine and Biotherapy (IRMB), CHU de Montpellier, Inserm U1183, université de Montpellier, Montpellier, France; Department of Cell and Tissue Engineering, CHU de Montpellier, University Montpellier, Montpellier, France
| |
Collapse
|
4
|
Dokukin NV, Chudakova DA, Shkap MO, Kovalchuk AM, Kibirsky PD, Baklaushev VP. Direct Neural Reprogramming in situ: Existing Approaches and Their Optimization. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:214-230. [PMID: 40254400 DOI: 10.1134/s000629792460426x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 04/22/2025]
Abstract
Direct in situ neuronal reprogramming (transdifferentiation) of glial cells (astrocytes and microglia) has attracted a significant interest as a potential approach for the treatment of a wide range of neurodegenerative diseases and damages of the central nervous system (CNS). The nervous system of higher mammals has a very limited capacity for repair. Disruption of CNS functioning due to traumatic injuries or neurodegenerative processes can significantly affect the quality of patients' life, lead to motor and cognitive impairments, and result in disability and, in some cases, death. Restoration of lost neurons in situ via direct reprogramming of glial cells without the intermediate stage of pluripotency seems to be the most attractive approach from the viewpoint of translational biomedicine. The ability of astroglia to actively proliferate in response to the damage of neural tissue supports the idea that these neuron-like cells, which are already present at the lesion site, are good candidates for transdifferentiation into neurons, considering that the possibility of direct neuronal reprogramming of astrocytes both in vitro and in vivo have demonstrated in many independent studies. Overexpression of proneuronal transcription factors, e.g., neurogenic differentiation factors 1-4 (NeuroD1-4), Neurogenin 2 (NeuroG2), Ascl1 (Achaete-Scute homolog 1), and Dlx2 (distal-less homeobox 2), including pioneer transcription factors that recognize target sequences in the compacted chromatin and activate transcription of silent genes, has already been proven as a potential therapeutic strategy. Other strategies, such as microRNA-mediated suppression of activity of PTB and REST transcription factors and application of small molecules or various biomaterials, are also utilized in neuronal reprogramming. However, the efficiency of direct in situ reprogramming is limited by a number of factors, including cell specificity of transgene delivery systems and promoters, brain regions in which transdifferentiation occurs, factors affecting cell metabolism, microenvironment, etc. Reprogramming in situ, which takes place in the presence of a large number of different cell types, requires monitoring and precise phenotypic characterization of subpopulations of cells undergoing transdifferentiation in order to confirm the reprogramming of the astroglia into neurons and subsequent integration of these neurons into the CNS. Here, we discussed the most efficient strategies of neuronal reprogramming and technologies used to visualize the transdifferentiation process, with special focus on the obstacles to efficient neuronal conversion, as well as approaches to overcome them.
Collapse
Affiliation(s)
- Nikita V Dokukin
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Daria A Chudakova
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
- National Medical Research Center of Children's Health, Ministry of Health of the Russian Federation, Moscow, 119991, Russia
| | - Matvey O Shkap
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Anna M Kovalchuk
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Pavel D Kibirsky
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Vladimir P Baklaushev
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia.
- Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, Moscow, 115682, Russia
- Research Institute of Pulmonology, Federal Medical and Biological Agency of Russia, Moscow, 115682, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| |
Collapse
|
5
|
Sun D, Chang Q, Lu F. Immunomodulation in diabetic wounds healing: The intersection of macrophage reprogramming and immunotherapeutic hydrogels. J Tissue Eng 2024; 15:20417314241265202. [PMID: 39071896 PMCID: PMC11283672 DOI: 10.1177/20417314241265202] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Diabetic wound healing presents a significant clinical challenge due to the interplay of systemic metabolic disturbances and local inflammation, which hinder the healing process. Macrophages undergo a phenotypic shift from M1 to M2 during wound healing, a transition pivotal for effective tissue repair. However, in diabetic wounds, the microenvironment disrupts this phenotypic polarization, perpetuating inflammation, and impeding healing. Reprograming macrophages to restore their M2 phenotype offers a potential avenue for modulating the wound immune microenvironment and promoting healing. This review elucidates the mechanisms underlying impaired macrophage polarization toward the M2 phenotype in diabetic wounds and discusses novel strategies, including epigenetic and metabolic interventions, to promote macrophage conversion to M2. Hydrogels, with their hydrated 3D cross-linked structure, closely resemble the physiological extracellular matrix and offer advantageous properties such as biocompatibility, tunability, and versatility. These characteristics make hydrogels promising candidates for developing immunomodulatory materials aimed at addressing diabetic wounds. Understanding the role of hydrogels in immunotherapy, particularly in the context of macrophage reprograming, is essential for the development of advanced wound care solutions. This review also highlights recent advancements in immunotherapeutic hydrogels as a step toward precise and effective treatments for diabetic wounds.
Collapse
Affiliation(s)
- Dan Sun
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Robinson EL, Ameri P, Delrue L, Vanderheyden M, Bartunek J, Altieri P, Heymans S, Heggermont WA. Differential expression of epigenetic modifiers in early and late cardiotoxic heart failure reveals DNA methylation as a key regulator of cardiotoxicity. Front Cardiovasc Med 2023; 10:884174. [PMID: 36970338 PMCID: PMC10034031 DOI: 10.3389/fcvm.2023.884174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
BackgroundAnthracycline-induced cardiotoxicity is a well-known serious clinical entity. However, detailed mechanistic insights on how short-term administration leads to late and long-lasting cardiotoxicity, are still largely undiscovered. We hypothesize that chemotherapy provokes a memory effect at the level of epigenomic DNA modifications which subsequently lead to cardiotoxicity even years after cessation of chemotherapy.MethodsWe explored the temporal evolution of epigenetic modifiers in early and late cardiotoxicity due to anthracyclines by means of RNA-sequencing of human endomyocardial left ventricular biopsies and mass spectrometry of genomic DNA. Based on these findings, validation of differentially regulated genes was obtained by performing RT-qPCR. Finally, a proof-of-concept in vitro mechanistic study was performed to dissect some of the mechanistic aspects of epigenetic memory in anthracycline-induced cardiotoxicity.ResultsCorrelation of gene expression between late and early onset cardiotoxicity revealed an R2 value of 0.98, demonstrating a total of 369 differentially expressed genes (DEGs, FDR < 0.05). of which 72% (n = 266) were upregulated, and 28% of genes, (n = 103) downregulated in later as compared to earlier onset cardiotoxicity. Gene ontology analysis showed significant enrichment of genes involved in methyl-CpG DNA binding, chromatin remodeling and regulation of transcription and positive regulation of apoptosis. Differential mRNA expression of genes involved in DNA methylation metabolism were confirmed by RT-qPCR in endomyocardial biopsies. In a larger biopsy cohort, it was shown that Tet2 was more abundantly expressed in cardiotoxicity biopsies vs. control biopsies and vs. non-ischemic cardiomyopathy patients. Moreover, an in vitro study was performed: following short-term doxorubicin treatment, H9c2 cells were cultured and passaged once they reached a confluency of 70%–80%. When compared to vehicle-only treated cells, in doxorubicin-treated cells, three weeks after short term treatment, Nppa, Nppb, Tet1/2 and other genes involved in active DNA demethylation were markedly upregulated. These alterations coincided with a loss of DNA methylation and a gain in hydroxymethylation, reflecting the epigenetic changes seen in the endomyocardial biopsies.ConclusionsShort-term administration of anthracyclines provokes long-lasting epigenetic modifications in cardiomyocytes both in vivo and in vitro, which explain in part the time lapse between the use of chemotherapy and the development of cardiotoxicity and, eventually, heart failure.
Collapse
Affiliation(s)
- Emma L. Robinson
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genova, Italy
- Cardiothoracic and Vascular Department, Ospedale Policlinico San Martino – IRCCS Italian Cardiovascular Network, Genova, Italy
| | - Leen Delrue
- Cardiovascular Research Center Aalst, Onze-Lieve-Vrouw Hospital Aalst, Aalst, Belgium
| | - Marc Vanderheyden
- Cardiovascular Research Center Aalst, Onze-Lieve-Vrouw Hospital Aalst, Aalst, Belgium
| | - Jozef Bartunek
- Cardiovascular Research Center Aalst, Onze-Lieve-Vrouw Hospital Aalst, Aalst, Belgium
| | - Paola Altieri
- Department of Internal Medicine, University of Genova, Genova, Italy
- Cardiothoracic and Vascular Department, Ospedale Policlinico San Martino – IRCCS Italian Cardiovascular Network, Genova, Italy
| | - Stephane Heymans
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Ward A. Heggermont
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Cardiovascular Research Center Aalst, Onze-Lieve-Vrouw Hospital Aalst, Aalst, Belgium
- Correspondence: Ward A. Heggermont
| |
Collapse
|
7
|
Araki H, Hino S, Anan K, Kuribayashi K, Etoh K, Seko D, Takase R, Kohrogi K, Hino Y, Ono Y, Araki E, Nakao M. LSD1 defines the fiber type-selective responsiveness to environmental stress in skeletal muscle. eLife 2023; 12:84618. [PMID: 36695573 PMCID: PMC9876571 DOI: 10.7554/elife.84618] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Skeletal muscle exhibits remarkable plasticity in response to environmental cues, with stress-dependent effects on the fast-twitch and slow-twitch fibers. Although stress-induced gene expression underlies environmental adaptation, it is unclear how transcriptional and epigenetic factors regulate fiber type-specific responses in the muscle. Here, we show that flavin-dependent lysine-specific demethylase-1 (LSD1) differentially controls responses to glucocorticoid and exercise in postnatal skeletal muscle. Using skeletal muscle-specific LSD1-knockout mice and in vitro approaches, we found that LSD1 loss exacerbated glucocorticoid-induced atrophy in the fast fiber-dominant muscles, with reduced nuclear retention of Foxk1, an anti-autophagic transcription factor. Furthermore, LSD1 depletion enhanced endurance exercise-induced hypertrophy in the slow fiber-dominant muscles, by induced expression of ERRγ, a transcription factor that promotes oxidative metabolism genes. Thus, LSD1 serves as an 'epigenetic barrier' that optimizes fiber type-specific responses and muscle mass under the stress conditions. Our results uncover that LSD1 modulators provide emerging therapeutic and preventive strategies against stress-induced myopathies such as sarcopenia, cachexia, and disuse atrophy.
Collapse
Affiliation(s)
- Hirotaka Araki
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto UniversityKumamotoJapan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kotaro Anan
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kanji Kuribayashi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kan Etoh
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Daiki Seko
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
- Department of Molecular Bone Biology, Graduate School of Biomedical Sciences, Nagasaki UniversityNagasakiJapan
| | - Ryuta Takase
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Kensaku Kohrogi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Yuko Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto UniversityKumamotoJapan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamotoJapan
| |
Collapse
|
8
|
Abstract
Cellular identity is established through complex layers of genetic regulation, forged over a developmental lifetime. An expanding molecular toolbox is allowing us to manipulate these gene regulatory networks in specific cell types in vivo. In principle, if we found the right molecular tricks, we could rewrite cell identity and harness the rich repertoire of possible cellular functions and attributes. Recent work suggests that this rewriting of cell identity is not only possible, but that newly induced cells can mitigate disease phenotypes in animal models of major human diseases. So, is the sky the limit, or do we need to keep our feet on the ground? This Spotlight synthesises key concepts emerging from recent efforts to reprogramme cellular identity in vivo. We provide our perspectives on recent controversies in the field of glia-to-neuron reprogramming and identify important gaps in our understanding that present barriers to progress.
Collapse
Affiliation(s)
- Sydney Leaman
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK
| | - Benedikt Berninger
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.,Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany.,The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|