1
|
Chou CC, Vest R, Prado MA, Wilson-Grady J, Paulo JA, Shibuya Y, Moran-Losada P, Lee TT, Luo J, Gygi SP, Kelly JW, Finley D, Wernig M, Wyss-Coray T, Frydman J. Proteostasis and lysosomal repair deficits in transdifferentiated neurons of Alzheimer's disease. Nat Cell Biol 2025; 27:619-632. [PMID: 40140603 PMCID: PMC11991917 DOI: 10.1038/s41556-025-01623-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 01/21/2025] [Indexed: 03/28/2025]
Abstract
Ageing is the most prominent risk factor for Alzheimer's disease (AD). However, the cellular mechanisms linking neuronal proteostasis decline to the characteristic aberrant protein deposits in the brains of patients with AD remain elusive. Here we develop transdifferentiated neurons (tNeurons) from human dermal fibroblasts as a neuronal model that retains ageing hallmarks and exhibits AD-linked vulnerabilities. Remarkably, AD tNeurons accumulate proteotoxic deposits, including phospho-tau and amyloid β, resembling those in APP mouse brains and the brains of patients with AD. Quantitative tNeuron proteomics identify ageing- and AD-linked deficits in proteostasis and organelle homeostasis, most notably in endosome-lysosomal components. Lysosomal deficits in aged tNeurons, including constitutive lysosomal damage and ESCRT-mediated lysosomal repair defects, are exacerbated in AD tNeurons and linked to inflammatory cytokine secretion and cell death. Providing support for the centrality of lysosomal deficits in AD, compounds ameliorating lysosomal function reduce amyloid β deposits and cytokine secretion. Thus, the tNeuron model system reveals impaired lysosomal homeostasis as an early event of ageing and AD.
Collapse
Affiliation(s)
- Ching-Chieh Chou
- Department of Biology, Stanford University, Stanford, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Qinotto Inc., San Carlos, CA, USA
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yohei Shibuya
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia Moran-Losada
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research Inc. (PAVIR), Palo Alto, CA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences and The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Cates K, Yuan L, Yang Y, Yoo AS. Fate erasure logic of gene networks underlying direct neuronal conversion of somatic cells by microRNAs. Cell Rep 2025; 44:115153. [PMID: 39756035 PMCID: PMC11834941 DOI: 10.1016/j.celrep.2024.115153] [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: 02/23/2024] [Revised: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/07/2025] Open
Abstract
Neurogenic microRNAs 9/9∗ and 124 (miR-9/9∗-124) drive the direct reprogramming of human fibroblasts into neurons with the initiation of the fate erasure of fibroblasts. However, whether the miR-9/9∗-124 fate erasure logic extends to the neuronal conversion of other somatic cell types remains unknown. Here, we uncover that miR-9/9∗-124 induces neuronal conversion of multiple cell types: dura fibroblasts, astrocytes, smooth muscle cells, and pericytes. We reveal the cell-type-specific and pan-somatic gene network erasure induced by miR-9/9∗-124, including cell cycle, morphology, and proteostasis gene networks. Leveraging these pan-somatic gene networks, we predict upstream regulators that may antagonize somatic fate erasure. Among the predicted regulators, we identify TP53 (p53), whose inhibition is sufficient to enhance neuronal conversion even in post-mitotic cells. This study extends miR-9/9∗-124 reprogramming to alternate somatic cells, reveals the pan-somatic gene network fate erasure logic of miR-9/9∗-124, and shows a neurogenic role for p53 inhibition in the miR-9/9∗-124 signaling cascade.
Collapse
Affiliation(s)
- Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Luorongxin Yuan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yan Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
3
|
Mishra P, Biesiada I, Gupta P, Ghavami S, Markowski J, Łos MJ. Unraveling the Complexity and Advancements of Transdifferentiation Technologies in the Biomedical Field and Their Potential Clinical Relevance. Arch Immunol Ther Exp (Warsz) 2025; 73:aite-2025-0001. [PMID: 39637369 DOI: 10.2478/aite-2025-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Chronic diseases such as cancer, autoimmunity, and organ failure currently depend on conventional pharmaceutical treatment, which may cause detrimental side effects in the long term. In this regard, cell-based therapy has emerged as a suitable alternative for treating these chronic diseases. Transdifferentiation technologies have evolved as a suitable therapeutic alternative that converts one differentiated somatic cell into another phenotype by using transcription factors (TFs), small molecules, or small, single-stranded, non-coding RNA molecules (miRNA). The transdifferentiation techniques rely on simple, fast, standardized, and versatile protocols with minimal chance of tumorigenicity and genotoxicity. However, there are still challenges and limitations that need to be addressed to enhance their clinical translation percentage in the near future. Taking this into account, we have delineated the features and strategies used in the transdifferentiation techniques. Then, we delved into different intermediate states that were attained during transdifferentiation. Advancements in transdifferentiation techniques in the field of tissue engineering, autoimmunity, and cancer therapy were dissected. Furthermore, limitations, challenges, and future perspectives are outlined in this review to provide a whole new picture of the transdifferentiation techniques. Advancements in molecular biology, interdisciplinary research, bioinformatics, and artificial intelligence will push the frontiers of this technology further to establish new avenues for biomedical research.
Collapse
Affiliation(s)
- Purusottam Mishra
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Izabella Biesiada
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Payal Gupta
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland
| | - Jarosław Markowski
- Department of Laryngology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
4
|
Ostermann PN, Evering TH. The impact of aging on HIV-1-related neurocognitive impairment. Ageing Res Rev 2024; 102:102513. [PMID: 39307316 DOI: 10.1016/j.arr.2024.102513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
Depending on the population studied, HIV-1-related neurocognitive impairment is estimated to impact up to half the population of people living with HIV (PLWH) despite the availability of combination antiretroviral therapy (cART). Various factors contribute to this neurocognitive impairment, which complicates our understanding of the molecular mechanisms involved. Biological aging has been implicated as one factor possibly impacting the development and progression of HIV-1-related neurocognitive impairment. This is increasingly important as the life expectancy of PLWH with virologic suppression on cART is currently projected to be similar to that of individuals not living with HIV. Based on our increasing understanding of the biological aging process on a cellular level, we aim to dissect possible interactions of aging- and HIV-1 infection-induced effects and their role in neurocognitive decline. Thus, we begin by providing a brief overview of the clinical aspects of HIV-1-related neurocognitive impairment and review the accumulating evidence implicating aging in its development (Part I). We then discuss potential interactions between aging-associated pathways and HIV-1-induced effects at the molecular level (Part II).
Collapse
Affiliation(s)
- Philipp Niklas Ostermann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Teresa Hope Evering
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
5
|
Samoilova EM, Chudakova DA, Dashinimaev EB, Snezhkina AV, Kudryashova OM, Lipatova AV, Soboleva AV, Vorob’yev PO, Valuev-Elliston VT, Zakirova NF, Ivanov AV, Baklaushev VP. A Snapshot of Early Transcriptional Changes Accompanying the Pro-Neural Phenotype Switch by NGN2, ASCL1, SOX2, and MSI1 in Human Fibroblasts: An RNA-Seq Study. Int J Mol Sci 2024; 25:12385. [PMID: 39596450 PMCID: PMC11594342 DOI: 10.3390/ijms252212385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Direct pro-neural reprogramming is a conversion of differentiated somatic cells to neural cells without an intermediate pluripotency stage. It is usually achieved via ectopic expression (EE) of certain transcription factors (TFs) or other reprogramming factors (RFs). Determining the transcriptional changes (TCs) caused by particular RFs in a given cell line enables an informed approach to reprogramming initiation. Here, we characterized TCs in the human fibroblast cell line LF1 on the 5th day after EE of the single well-known pro-neural RFs NGN2, ASCL1, SOX2, and MSI1. As assessed by expression analysis of the bona fide neuronal markers nestin and beta-III tubulin, all four RFs initiated pro-neuronal phenotype conversion; analysis by RNA-seq revealed striking differences in the resulting TCs, although some pathways were overlapping. ASCL1 and SOX2 were not sufficient to induce significant pro-neural phenotype switches using our EE system. NGN2 induced TCs indicative of cell phenotype changes towards neural crest cells, neural stem cells, mature neurons, as well as radial glia, astrocytes, and oligodendrocyte precursors and their mature forms. MSI1 mainly induced a switch towards early stem-like cells, such as radial glia.
Collapse
Affiliation(s)
- Ekaterina M. Samoilova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- National Medical Research Center of Children’s Health of the Ministry of Health of the Russian Federation, 119296 Moscow, Russia
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Olga M. Kudryashova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Alesya V. Soboleva
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Pavel O. Vorob’yev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Vladimir T. Valuev-Elliston
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Natalia F. Zakirova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia; (E.M.S.); (O.M.K.); (A.V.S.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.V.S.); (A.V.L.); (P.O.V.); (V.T.V.-E.); (A.V.I.)
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Pulmonology Research Institute, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| |
Collapse
|
6
|
Ostermann PN, Wu Y, Bowler SA, Siddiqui MA, Herrera A, Sidharta M, Ramnarine K, Martínez-Meza S, St. Bernard LA, Nixon DF, Jones RB, Yamashita M, Ndhlovu LC, Zhou T, Evering TH. A Transcriptional Signature of Induced Neurons Differentiates Virologically Suppressed People Living With HIV from People Without HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619617. [PMID: 39484396 PMCID: PMC11526917 DOI: 10.1101/2024.10.22.619617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Neurocognitive impairment is a prevalent and important co-morbidity in virologically suppressed people living with HIV (PLWH), yet the underlying mechanisms remain elusive and treatments lacking. Here, we explored for the first time, use of participant-derived directly induced neurons (iNs) to model neuronal biology and injury in PLWH. iNs retain age- and disease-related features of the donors, providing unique opportunities to reveal novel aspects of neurological disorders. We obtained primary dermal fibroblasts from six virologically suppressed PLWH (range: 27 - 64 years, median: 53); 83% Male; 50% White) and seven matched people without HIV (PWOH) (range: 27 - 66, median: 55); 71% Male; 57% White). iNs were generated using transcription factors NGN2 and ASCL1, and validated by immunocytochemistry and single-cell-RNAseq. Transcriptomic analysis using bulk-RNAseq identified 29 significantly differentially expressed genes between iNs from PLWH and PWOH. Of these, 16 genes were downregulated and 13 upregulated in PLWH iNs. Protein-protein interaction network mapping indicates that iNs from PLWH exhibit differences in extracellular matrix organization and synaptic transmission. IFI27 was upregulated in iNs from PLWH, which complements independent post-mortem studies demonstrating elevated IFI27 expression in PLWH-derived brain tissue, indicating that iN generation reconstitutes this pathway. Finally, we observed that expression of the FOXL2NB-FOXL2-LINC01391 genome locus is reduced in iNs from PLWH and negatively correlates with neurocognitive impairment. Thus, we have identified an iN gene signature of HIV through direct reprogramming of skin fibroblasts into neurons revealing novel mechanisms of neurocognitive impairment in PLWH.
Collapse
Affiliation(s)
- Philipp N. Ostermann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Youjun Wu
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Scott A. Bowler
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mohammad Adnan Siddiqui
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY. 10032, USA
| | - Alberto Herrera
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mega Sidharta
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Kiran Ramnarine
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Samuel Martínez-Meza
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Leslie Ann St. Bernard
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Douglas F. Nixon
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - R. Brad Jones
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Masahiro Yamashita
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY. 10032, USA
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ting Zhou
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Teresa H. Evering
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
7
|
Chou CC, Vest R, Prado MA, Wilson-Grady J, Paulo JA, Shibuya Y, Moran-Losada P, Lee TT, Luo J, Gygi SP, Kelly JW, Finley D, Wernig M, Wyss-Coray T, Frydman J. Human tNeurons reveal aging-linked proteostasis deficits driving Alzheimer's phenotypes. RESEARCH SQUARE 2024:rs.3.rs-4407236. [PMID: 38853828 PMCID: PMC11160905 DOI: 10.21203/rs.3.rs-4407236/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Aging is a prominent risk factor for Alzheimer's disease (AD), but the cellular mechanisms underlying neuronal phenotypes remain elusive. Both accumulation of amyloid plaques and neurofibrillary tangles in the brain1 and age-linked organelle deficits2-7 are proposed as causes of AD phenotypes but the relationship between these events is unclear. Here, we address this question using a transdifferentiated neuron (tNeuron) model directly from human dermal fibroblasts. Patient-derived tNeurons retain aging hallmarks and exhibit AD-linked deficits. Quantitative tNeuron proteomic analyses identify aging and AD-linked deficits in proteostasis and organelle homeostasis, particularly affecting endosome-lysosomal components. The proteostasis and lysosomal homeostasis deficits in aged tNeurons are exacerbated in sporadic and familial AD tNeurons, promoting constitutive lysosomal damage and defects in ESCRT-mediated repair. We find deficits in neuronal lysosomal homeostasis lead to inflammatory cytokine secretion, cell death and spontaneous development of Aß and phospho-Tau deposits. These proteotoxic inclusions co-localize with lysosomes and damage markers and resemble inclusions in brain tissue from AD patients and APP-transgenic mice. Supporting the centrality of lysosomal deficits driving AD phenotypes, lysosome-function enhancing compounds reduce AD-associated cytokine secretion and Aβ deposits. We conclude that proteostasis and organelle deficits are upstream initiating factors leading to neuronal aging and AD phenotypes.
Collapse
Affiliation(s)
- Ching-Chieh Chou
- Department of Biology, Stanford University, Stanford, California, USA
| | - Ryan Vest
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
- Qinotto, Inc. San Carlos, California, USA
| | - Miguel A. Prado
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Joshua Wilson-Grady
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yohei Shibuya
- Departments of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Patricia Moran-Losada
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Ting-Ting Lee
- Department of Biology, Stanford University, Stanford, California, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, Inc. (PAVIR), Palo Alto, California, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, California, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Marius Wernig
- Departments of Pathology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California, USA
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, California, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Lead contact
| |
Collapse
|
8
|
Zardetto B, van Roon-Mom W, Aartsma-Rus A, Lauffer MC. Treatability of the KMT2-Associated Neurodevelopmental Disorders Using Antisense Oligonucleotide-Based Treatments. Hum Mutat 2024; 2024:9933129. [PMID: 40225946 PMCID: PMC11925151 DOI: 10.1155/2024/9933129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 04/15/2025]
Abstract
Neurodevelopmental disorders (NDDs) of genetic origin are a group of early-onset neurological diseases with highly heterogeneous etiology and a symptomatic spectrum that includes intellectual disability, autism spectrum disorder, and learning and language disorders. One group of rare NDDs is associated with dysregulation of the KMT2 protein family. Members of this family share a common methyl transferase function and are involved in the etiology of rare haploinsufficiency disorders. For each of the KMT2 genes, at least one distinct disorder has been reported, yet clinical manifestations often overlap for multiple of these individually very rare disorders. Clinical care is currently focused on the management of symptoms with no targeted treatments available, illustrating a high unmet medical need and the urgency of developing disease-modifying therapeutic strategies. Antisense oligonucleotides (ASOs) are one option to treat some of these rare genetic disorders. ASOs are RNA-based treatments that can be employed to modulate gene expression through various mechanisms. In this work, we discuss the phenotypic features across the KMT2-associated NDDs and which ASO approaches are most suited for the treatment of each associated disorder. We hereby address variant-specific strategies as well as options applicable to larger groups of patients.
Collapse
Affiliation(s)
- Bianca Zardetto
- Dutch Center for RNA TherapeuticsDepartment of Human GeneticsLeiden University Medical CenterLeiden, Netherlands
| | - Willeke van Roon-Mom
- Dutch Center for RNA TherapeuticsDepartment of Human GeneticsLeiden University Medical CenterLeiden, Netherlands
| | - Annemieke Aartsma-Rus
- Dutch Center for RNA TherapeuticsDepartment of Human GeneticsLeiden University Medical CenterLeiden, Netherlands
| | - Marlen C. Lauffer
- Dutch Center for RNA TherapeuticsDepartment of Human GeneticsLeiden University Medical CenterLeiden, Netherlands
| |
Collapse
|
9
|
Li S, Zhao S, Sinson JC, Bajic A, Rosenfeld JA, Neeley MB, Pena M, Worley KC, Burrage LC, Weisz-Hubshman M, Ketkar S, Craigen WJ, Clark GD, Lalani S, Bacino CA, Machol K, Chao HT, Potocki L, Emrick L, Sheppard J, Nguyen MTT, Khoramnia A, Hernandez PP, Nagamani SC, Liu Z, Eng CM, Lee B, Liu P. The clinical utility and diagnostic implementation of human subject cell transdifferentiation followed by RNA sequencing. Am J Hum Genet 2024; 111:841-862. [PMID: 38593811 PMCID: PMC11080285 DOI: 10.1016/j.ajhg.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
RNA sequencing (RNA-seq) has recently been used in translational research settings to facilitate diagnoses of Mendelian disorders. A significant obstacle for clinical laboratories in adopting RNA-seq is the low or absent expression of a significant number of disease-associated genes/transcripts in clinically accessible samples. As this is especially problematic in neurological diseases, we developed a clinical diagnostic approach that enhanced the detection and evaluation of tissue-specific genes/transcripts through fibroblast-to-neuron cell transdifferentiation. The approach is designed specifically to suit clinical implementation, emphasizing simplicity, cost effectiveness, turnaround time, and reproducibility. For clinical validation, we generated induced neurons (iNeurons) from 71 individuals with primary neurological phenotypes recruited to the Undiagnosed Diseases Network. The overall diagnostic yield was 25.4%. Over a quarter of the diagnostic findings benefited from transdifferentiation and could not be achieved by fibroblast RNA-seq alone. This iNeuron transcriptomic approach can be effectively integrated into diagnostic whole-transcriptome evaluation of individuals with genetic disorders.
Collapse
Affiliation(s)
- Shenglan Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Sen Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jefferson C Sinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Aleksandar Bajic
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Advanced Technology Cores, Baylor College of Medicine, Houston, TX, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Matthew B Neeley
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA
| | - Mezthly Pena
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Kim C Worley
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Monika Weisz-Hubshman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Shamika Ketkar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Gary D Clark
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Seema Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Carlos A Bacino
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Keren Machol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Hsiao-Tuan Chao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Cain Pediatric Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, USA
| | - Lorraine Potocki
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Lisa Emrick
- Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Jennifer Sheppard
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - My T T Nguyen
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Anahita Khoramnia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Sandesh Cs Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Christine M Eng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Baylor Genetics, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Baylor Genetics, Houston, TX, USA.
| |
Collapse
|
10
|
Harley J, Santosa MM, Ng CY, Grinchuk OV, Hor JH, Liang Y, Lim VJ, Tee WW, Ong DST, Ng SY. Telomere shortening induces aging-associated phenotypes in hiPSC-derived neurons and astrocytes. Biogerontology 2024; 25:341-360. [PMID: 37987889 PMCID: PMC10998800 DOI: 10.1007/s10522-023-10076-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/13/2023] [Indexed: 11/22/2023]
Abstract
Telomere shortening is a well-established hallmark of cellular aging. Telomerase reverse transcriptase (TERT) plays a crucial role in maintaining the length of telomeres, which are specialised protective caps at the end of chromosomes. The lack of in vitro aging models, particularly for the central nervous system (CNS), has impeded progress in understanding aging and age-associated neurodegenerative diseases. In this study, we aimed to explore the possibility of inducing aging-associated features in cell types of the CNS using hiPSC (human induced pluripotent stem cell) technology. To achieve this, we utilised CRISPR/Cas9 to generate hiPSCs with a loss of telomerase function and shortened telomeres. Through directed differentiation, we generated motor neurons and astrocytes to investigate whether telomere shortening could lead to age-associated phenotypes. Our findings revealed that shortened telomeres induced age-associated characteristics in both motor neurons and astrocytes including increased cellular senescence, heightened inflammation, and elevated DNA damage. We also observed cell-type specific age-related morphology changes. Additionally, our study highlighted the fundamental role of TERT and telomere shortening in neural progenitor cell (NPC) proliferation and neuronal differentiation. This study serves as a proof of concept that telomere shortening can effectively induce aging-associated phenotypes, thereby providing a valuable tool to investigate age-related decline and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jasmine Harley
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Munirah Mohamad Santosa
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Chong Yi Ng
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Oleg V Grinchuk
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Jin-Hui Hor
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Yajing Liang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Valerie Jingwen Lim
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
| | - Wee Wei Tee
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Derrick Sek Tong Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Shi-Yan Ng
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore, 138673, Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
11
|
Marzoog BA. Transcription Factors in Brain Regeneration: A Potential Novel Therapeutic Target. Curr Drug Targets 2024; 25:46-61. [PMID: 38444255 DOI: 10.2174/0113894501279977231210170231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 03/07/2024]
Abstract
Transcription factors play a crucial role in providing identity to each cell population. To maintain cell identity, it is essential to balance the expression of activator and inhibitor transcription factors. Cell plasticity and reprogramming offer great potential for future therapeutic applications, as they can regenerate damaged tissue. Specific niche factors can modify gene expression and differentiate or transdifferentiate the target cell to the required fate. Ongoing research is being carried out on the possibilities of transcription factors in regenerating neurons, with neural stem cells (NSCs) being considered the preferred cells for generating new neurons due to their epigenomic and transcriptome memory. NEUROD1/ASCL1, BRN2, MYTL1, and other transcription factors can induce direct reprogramming of somatic cells, such as fibroblasts, into neurons. However, the molecular biology of transcription factors in reprogramming and differentiation still needs to be fully understood.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- World-Class Research Center, Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
12
|
Gomez Ramos B, Ohnmacht J, de Lange N, Valceschini E, Ginolhac A, Catillon M, Ferrante D, Rakovic A, Halder R, Massart F, Arena G, Antony P, Bolognin S, Klein C, Krause R, Schulz MH, Sauter T, Krüger R, Sinkkonen L. Multiomics analysis identifies novel facilitators of human dopaminergic neuron differentiation. EMBO Rep 2024; 25:254-285. [PMID: 38177910 PMCID: PMC10897179 DOI: 10.1038/s44319-023-00024-2] [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: 04/17/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 01/06/2024] Open
Abstract
Midbrain dopaminergic neurons (mDANs) control voluntary movement, cognition, and reward behavior under physiological conditions and are implicated in human diseases such as Parkinson's disease (PD). Many transcription factors (TFs) controlling human mDAN differentiation during development have been described, but much of the regulatory landscape remains undefined. Using a tyrosine hydroxylase (TH) human iPSC reporter line, we here generate time series transcriptomic and epigenomic profiles of purified mDANs during differentiation. Integrative analysis predicts novel regulators of mDAN differentiation and super-enhancers are used to identify key TFs. We find LBX1, NHLH1 and NR2F1/2 to promote mDAN differentiation and show that overexpression of either LBX1 or NHLH1 can also improve mDAN specification. A more detailed investigation of TF targets reveals that NHLH1 promotes the induction of neuronal miR-124, LBX1 regulates cholesterol biosynthesis, and NR2F1/2 controls neuronal activity.
Collapse
Affiliation(s)
- Borja Gomez Ramos
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Jochen Ohnmacht
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Nikola de Lange
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Elena Valceschini
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Marie Catillon
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Daniele Ferrante
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Aleksandar Rakovic
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - François Massart
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Giuseppe Arena
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Paul Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Silvia Bolognin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, 23538, Lübeck, Germany
| | - Roland Krause
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Marcel H Schulz
- Institute for Cardiovascular Regeneration, Goethe University, 60590, Frankfurt, Germany
- German Centre for Cardiovascular Research, Partner site Rhein-Main, 60590, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, Goethe University, Frankfurt am Main, Germany
| | - Thomas Sauter
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362, Belvaux, Luxembourg
- Centre Hospitalier de Luxembourg (CHL), L-1210, Luxembourg, Luxembourg
- Luxembourg Institute of Health (LIH), L-1445, Luxembourg, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, Belvaux, Luxembourg.
| |
Collapse
|
13
|
Chudakova DA, Samoilova EM, Chekhonin VP, Baklaushev VP. Improving Efficiency of Direct Pro-Neural Reprogramming: Much-Needed Aid for Neuroregeneration in Spinal Cord Injury. Cells 2023; 12:2499. [PMID: 37887343 PMCID: PMC10605572 DOI: 10.3390/cells12202499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Spinal cord injury (SCI) is a medical condition affecting ~2.5-4 million people worldwide. The conventional therapy for SCI fails to restore the lost spinal cord functions; thus, novel therapies are needed. Recent breakthroughs in stem cell biology and cell reprogramming revolutionized the field. Of them, the use of neural progenitor cells (NPCs) directly reprogrammed from non-neuronal somatic cells without transitioning through a pluripotent state is a particularly attractive strategy. This allows to "scale up" NPCs in vitro and, via their transplantation to the lesion area, partially compensate for the limited regenerative plasticity of the adult spinal cord in humans. As recently demonstrated in non-human primates, implanted NPCs contribute to the functional improvement of the spinal cord after injury, and works in other animal models of SCI also confirm their therapeutic value. However, direct reprogramming still remains a challenge in many aspects; one of them is low efficiency, which prevents it from finding its place in clinics yet. In this review, we describe new insights that recent works brought to the field, such as novel targets (mitochondria, nucleoli, G-quadruplexes, and others), tools, and approaches (mechanotransduction and electrical stimulation) for direct pro-neural reprogramming, including potential ones yet to be tested.
Collapse
Affiliation(s)
- Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Ekaterina M. Samoilova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialised Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialised Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| |
Collapse
|
14
|
Ostermann PN, Schaal H. Human brain organoids to explore SARS-CoV-2-induced effects on the central nervous system. Rev Med Virol 2023; 33:e2430. [PMID: 36790825 DOI: 10.1002/rmv.2430] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/16/2023]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). In less than three years, an estimated 600 million infections with SARS-CoV-2 occurred worldwide, resulting in a pandemic with tremendous impact especially on economic and health sectors. Initially considered a respiratory disease, COVID-19, along with its long-term sequelae (long-COVID) rather is a systemic disease. Neurological symptoms like dementia or encephalopathy were reported early during the pandemic as concomitants of the acute phase and as characteristics of long-COVID. An excessive inflammatory immune response is hypothesized to play a major role in this context. However, direct infection of neural cells may also contribute to the neurological aspects of (long)-COVID-19. To mainly explore such direct effects of SARS-CoV-2 on the central nervous system, human brain organoids provide a useful platform. Infecting these three-dimensional tissue cultures allows the study of viral neurotropism as well as of virus-induced effects on single cells or even the complex cellular network within the organoid. In this review, we summarize the experimental studies that used SARS-CoV-2-infected human brain organoids to unravel the complex nature of (long)-COVID-19-related neurological manifestations.
Collapse
Affiliation(s)
- Philipp Niklas Ostermann
- Institute of Virology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Heiner Schaal
- Institute of Virology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
15
|
Herdy JR, Traxler L, Agarwal RK, Karbacher L, Schlachetzki JCM, Boehnke L, Zangwill D, Galasko D, Glass CK, Mertens J, Gage FH. Increased post-mitotic senescence in aged human neurons is a pathological feature of Alzheimer's disease. Cell Stem Cell 2022; 29:1637-1652.e6. [PMID: 36459967 PMCID: PMC10093780 DOI: 10.1016/j.stem.2022.11.010] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 10/03/2022] [Accepted: 11/09/2022] [Indexed: 12/02/2022]
Abstract
The concept of senescence as a phenomenon limited to proliferating cells has been challenged by growing evidence of senescence-like features in terminally differentiated cells, including neurons. The persistence of senescent cells late in life is associated with tissue dysfunction and increased risk of age-related disease. We found that Alzheimer's disease (AD) brains have significantly higher proportions of neurons that express senescence markers, and their distribution indicates bystander effects. AD patient-derived directly induced neurons (iNs) exhibit strong transcriptomic, epigenetic, and molecular biomarker signatures, indicating a specific human neuronal senescence-like state. AD iN single-cell transcriptomics revealed that senescent-like neurons face oncogenic challenges and metabolic dysfunction as well as display a pro-inflammatory signature. Integrative profiling of the inflammatory secretome of AD iNs and patient cerebral spinal fluid revealed a neuronal senescence-associated secretory phenotype that could trigger astrogliosis in human astrocytes. Finally, we show that targeting senescence-like neurons with senotherapeutics could be a strategy for preventing or treating AD.
Collapse
Affiliation(s)
- Joseph R Herdy
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria.
| | - Larissa Traxler
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Ravi K Agarwal
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lukas Karbacher
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Lena Boehnke
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Dina Zangwill
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Doug Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jerome Mertens
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria.
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
16
|
Traxler L, Herdy JR, Stefanoni D, Eichhorner S, Pelucchi S, Szücs A, Santagostino A, Kim Y, Agarwal RK, Schlachetzki JCM, Glass CK, Lagerwall J, Galasko D, Gage FH, D'Alessandro A, Mertens J. Warburg-like metabolic transformation underlies neuronal degeneration in sporadic Alzheimer's disease. Cell Metab 2022; 34:1248-1263.e6. [PMID: 35987203 PMCID: PMC9458870 DOI: 10.1016/j.cmet.2022.07.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 04/13/2022] [Accepted: 07/26/2022] [Indexed: 12/28/2022]
Abstract
The drivers of sporadic Alzheimer's disease (AD) remain incompletely understood. Utilizing directly converted induced neurons (iNs) from AD-patient-derived fibroblasts, we identified a metabolic switch to aerobic glycolysis in AD iNs. Pathological isoform switching of the glycolytic enzyme pyruvate kinase M (PKM) toward the cancer-associated PKM2 isoform conferred metabolic and transcriptional changes in AD iNs. These alterations occurred via PKM2's lack of metabolic activity and via nuclear translocation and association with STAT3 and HIF1α to promote neuronal fate loss and vulnerability. Chemical modulation of PKM2 prevented nuclear translocation, restored a mature neuronal metabolism, reversed AD-specific gene expression changes, and re-activated neuronal resilience against cell death.
Collapse
Affiliation(s)
- Larissa Traxler
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria.
| | - Joseph R Herdy
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Davide Stefanoni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sophie Eichhorner
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria
| | - Silvia Pelucchi
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria
| | - Attila Szücs
- Neuronal Cell Biology Research Group, Eötvös Loránd University, Budapest 1117, Hungary
| | - Alice Santagostino
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria
| | - Yongsung Kim
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-5624, USA
| | - Ravi K Agarwal
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Jessica Lagerwall
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jerome Mertens
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University, Innsbruck 6020, Austria; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Böhnke L, Zhou-Yang L, Pelucchi S, Kogler F, Frantal D, Schön F, Lagerström S, Borgogno O, Baltazar J, Herdy JR, Kittel-Schneider S, Defrancesco M, Mertens J. Chemical Replacement of Noggin with Dorsomorphin Homolog 1 for Cost-Effective Direct Neuronal Conversion. Cell Reprogram 2022; 24:304-313. [PMID: 35877103 PMCID: PMC9587801 DOI: 10.1089/cell.2021.0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The direct conversion of adult human skin fibroblasts (FBs) into induced neurons (iNs) represents a useful technology to generate donor-specific adult-like human neurons. Disease modeling studies rely on the consistently efficient conversion of relatively large cohorts of FBs. Despite the identification of several small molecular enhancers, high-yield protocols still demand addition of recombinant Noggin. To identify a replacement to circumvent the technical and economic challenges associated with Noggin, we assessed dynamic gene expression trajectories of transforming growth factor-β signaling during FB-to-iN conversion. We identified ALK2 (ACVR1) of the bone morphogenic protein branch to possess the highest initial transcript abundance in FBs and the steepest decline during successful neuronal conversion. We thus assessed the efficacy of dorsomorphin homolog 1 (DMH1), a highly selective ALK2-inhibitor, for its potential to replace Noggin. Conversion media containing DMH1 (+DMH1) indeed enhanced conversion efficiencies over basic SMAD inhibition (tSMADi), yielding similar βIII-tubulin (TUBB3) purities as conversion media containing Noggin (+Noggin). Furthermore, +DMH1 induced high yields of iNs with clear neuronal morphologies that are positive for the mature neuronal marker NeuN. Validation of +DMH1 for iN conversion of FBs from 15 adult human donors further demonstrates that Noggin-free conversion consistently yields iN cultures that display high βIII-tubulin numbers with synaptic structures and basic spontaneous neuronal activity at a third of the cost.
Collapse
Affiliation(s)
- Lena Böhnke
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria.,Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Lucia Zhou-Yang
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Silvia Pelucchi
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Flora Kogler
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Daniela Frantal
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Florian Schön
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Stina Lagerström
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Oliver Borgogno
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Jennifer Baltazar
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Joseph R Herdy
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria.,Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Sarah Kittel-Schneider
- Center of Mental Health, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Würzburg, Würzburg, Bavaria, Germany
| | - Michaela Defrancesco
- Division of Psychiatry I, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Tyrol, Austria
| | - Jerome Mertens
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, University of Innsbruck, Innsbruck, Tyrol, Austria.,Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
18
|
Wang J, Chen S, Pan C, Li G, Tang Z. Application of Small Molecules in the Central Nervous System Direct Neuronal Reprogramming. Front Bioeng Biotechnol 2022; 10:799152. [PMID: 35875485 PMCID: PMC9301571 DOI: 10.3389/fbioe.2022.799152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
The lack of regenerative capacity of neurons leads to poor prognoses for some neurological disorders. The use of small molecules to directly reprogram somatic cells into neurons provides a new therapeutic strategy for neurological diseases. In this review, the mechanisms of action of different small molecules, the approaches to screening small molecule cocktails, and the methods employed to detect their reprogramming efficiency are discussed, and the studies, focusing on neuronal reprogramming using small molecules in neurological disease models, are collected. Future research efforts are needed to investigate the in vivo mechanisms of small molecule-mediated neuronal reprogramming under pathophysiological states, optimize screening cocktails and dosing regimens, and identify safe and effective delivery routes to promote neural regeneration in different neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Gaigai Li
- *Correspondence: Gaigai Li, ; Zhouping Tang,
| | | |
Collapse
|
19
|
Impact of Cultured Neuron Models on α-Herpesvirus Latency Research. Viruses 2022; 14:v14061209. [PMID: 35746680 PMCID: PMC9228292 DOI: 10.3390/v14061209] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
A signature trait of neurotropic α-herpesviruses (α-HV) is their ability to establish stable non-productive infections of peripheral neurons termed latency. This specialized gene expression program is the foundation of an evolutionarily successful strategy to ensure lifelong persistence in the host. Various physiological stresses can induce reactivation in a subset of latently-infected neurons allowing a new cycle of viral productive cycle gene expression and synthesis of infectious virus. Recurring reactivation events ensure transmission of the virus to new hosts and contributes to pathogenesis. Efforts to define the molecular basis of α-HV latency and reactivation have been notoriously difficult because the neurons harboring latent virus in humans and in experimentally infected live-animal models, are rare and largely inaccessible to study. Increasingly, researchers are turning to cultured neuron infection models as simpler experimental platforms from which to explore latency and reactivation at the molecular level. In this review, I reflect on the strengths and weaknesses of existing neuronal models and briefly summarize the important mechanistic insights these models have provided. I also discuss areas where prioritization will help to ensure continued progress and integration.
Collapse
|
20
|
Nishimura K, Nitta T, Doi K, Takata K. Rapid conversion of human induced pluripotent stem cells into dopaminergic neurons by inducible expression of two transcription factors. Stem Cells Dev 2022; 31:269-277. [PMID: 35420042 DOI: 10.1089/scd.2021.0363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) including human embryonic stem cells and human induced pluripotent stem cells (hiPSCs) provide promising sources for regenerative therapy, disease modeling, and drug screening. Relevant efforts have been invested in establishing robust induction protocols for PSC-derived dopaminergic (DA) neuron generation by mimicking brain development-related signaling pathways. However, these protocols require fully trained techniques and a long time to yield mature DA neurons. In this study, to accelerate the entire process, we generated a hiPSC line differentiating into DA neurons by the inducible force expression of two transcription factors ASCL1 and LMX1A. Using this hiPSC line, we established a rapid and simple induction protocol to generate mature DA neurons in 28 days. The induced DA neurons were characterized by gene expression and immunohistochemical analyses of fundamental DA neuronal markers. Moreover, the cell functional properties were analyzed by a multielectrode array system on day 28. This resource offers future applications for high-throughput screening, such as drug development and toxicology that require highly validated DA neurons.
Collapse
Affiliation(s)
- Kaneyasu Nishimura
- Kyoto Pharmaceutical University, 12916, Kyoto, Japan.,Doshisha University, 12757, Kyoto, Kyoto, Japan;
| | - Tatsumi Nitta
- Kyoto Pharmaceutical University, 12916, Kyoto, Kyoto, Japan;
| | - Keisuke Doi
- Kyoto Pharmaceutical University, 12916, Kyoto, Kyoto, Japan;
| | - Kazuyuki Takata
- Kyoto Pharmaceutical University, 12916, Kyoto, Kyoto, Japan;
| |
Collapse
|
21
|
Pircs K, Drouin-Ouellet J, Horváth V, Gil J, Rezeli M, Garza R, Grassi DA, Sharma Y, St-Amour I, Harris K, Jönsson ME, Johansson PA, Vuono R, Fazal SV, Stoker T, Hersbach BA, Sharma K, Lagerwall J, Lagerström S, Storm P, Hébert SS, Marko-Varga G, Parmar M, Barker RA, Jakobsson J. Distinct subcellular autophagy impairments in induced neurons from Huntington’s disease patients. Brain 2021; 145:3035-3057. [PMID: 34936701 PMCID: PMC9473361 DOI: 10.1093/brain/awab473] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 11/07/2021] [Accepted: 12/01/2021] [Indexed: 12/09/2022] Open
Abstract
Huntington's disease is a neurodegenerative disorder caused by CAG expansions in the huntingtin (HTT) gene. Modelling Huntington's disease is challenging, as rodent and cellular models poorly recapitulate the disease as seen in ageing humans. To address this, we generated induced neurons through direct reprogramming of human skin fibroblasts, which retain age-dependent epigenetic characteristics. Huntington's disease induced neurons (HD-iNs) displayed profound deficits in autophagy, characterized by reduced transport of late autophagic structures from the neurites to the soma. These neurite-specific alterations in autophagy resulted in shorter, thinner and fewer neurites specifically in HD-iNs. CRISPRi-mediated silencing of HTT did not rescue this phenotype but rather resulted in additional autophagy alterations in control induced neurons, highlighting the importance of wild-type HTT in normal neuronal autophagy. In summary, our work identifies a distinct subcellular autophagy impairment in adult patient derived Huntington's disease neurons and provides a new rationale for future development of autophagy activation therapies.
Collapse
Affiliation(s)
- Karolina Pircs
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Janelle Drouin-Ouellet
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, H3 T 1J4, Canada
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11 and B10, Lund University, S-221 84, Lund, Sweden
| | - Vivien Horváth
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Jeovanis Gil
- Oncology and Pathology, Kamprad Lab, Department of Clinical Sciences, Lund University, S-221 85, Lund, Sweden
| | - Melinda Rezeli
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, Lund University, S-221 85, Lund, Sweden
| | - Raquel Garza
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Daniela A. Grassi
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Yogita Sharma
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Isabelle St-Amour
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, CHUL, Québec, QC G1E 6W2, Canada
- CERVO Brain Research Center – Université Laval, Québec, QC G1E 1T2, Canada
| | - Kate Harris
- Wellcome-MRC Cambridge Stem Cell Institute & John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK
| | - Marie E. Jönsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Pia A. Johansson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Romina Vuono
- Wellcome-MRC Cambridge Stem Cell Institute & John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK
| | - Shaline V. Fazal
- Wellcome-MRC Cambridge Stem Cell Institute & John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK
| | - Thomas Stoker
- Wellcome-MRC Cambridge Stem Cell Institute & John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK
| | - Bob A. Hersbach
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Kritika Sharma
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Jessica Lagerwall
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Stina Lagerström
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| | - Petter Storm
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11 and B10, Lund University, S-221 84, Lund, Sweden
| | - Sébastien S. Hébert
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, CHUL, Québec, QC G1E 6W2, Canada
| | - György Marko-Varga
- Oncology and Pathology, Kamprad Lab, Department of Clinical Sciences, Lund University, S-221 85, Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11 and B10, Lund University, S-221 84, Lund, Sweden
| | - Roger A. Barker
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11 and B10, Lund University, S-221 84, Lund, Sweden
- Wellcome-MRC Cambridge Stem Cell Institute & John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK
| | - Johan Jakobsson
- Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, S-221 84, Lund, Sweden
| |
Collapse
|
22
|
Direct neuronal reprogramming: Fast forward from new concepts toward therapeutic approaches. Neuron 2021; 110:366-393. [PMID: 34921778 DOI: 10.1016/j.neuron.2021.11.023] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/25/2021] [Accepted: 11/19/2021] [Indexed: 12/21/2022]
Abstract
Differentiated cells have long been considered fixed in their identity. However, about 20 years ago, the first direct conversion of glial cells into neurons in vitro opened the field of "direct neuronal reprogramming." Since then, neuronal reprogramming has achieved the generation of fully functional, mature neurons with remarkable efficiency, even in diseased brain environments. Beyond their clinical implications, these discoveries provided basic insights into crucial mechanisms underlying conversion of specific cell types into neurons and maintenance of neuronal identity. Here we discuss such principles, including the importance of the starter cell for shaping the outcome of neuronal reprogramming. We further highlight technical concerns for in vivo reprogramming and propose a code of conduct to avoid artifacts and pitfalls. We end by pointing out next challenges for development of less invasive cell replacement therapies for humans.
Collapse
|
23
|
Han F, Liu Y, Huang J, Zhang X, Wei C. Current Approaches and Molecular Mechanisms for Directly Reprogramming Fibroblasts Into Neurons and Dopamine Neurons. Front Aging Neurosci 2021; 13:738529. [PMID: 34658841 PMCID: PMC8515543 DOI: 10.3389/fnagi.2021.738529] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease is mainly caused by specific degeneration of dopaminergic neurons (DA neurons) in the substantia nigra of the middle brain. Over the past two decades, transplantation of neural stem cells (NSCs) from fetal brain-derived neural stem cells (fNSCs), human embryonic stem cells (hESCs), and induced pluripotent stem cells (iPSCs) has been shown to improve the symptoms of motor dysfunction in Parkinson's disease (PD) animal models and PD patients significantly. However, there are ethical concerns with fNSCs and hESCs and there is an issue of rejection by the immune system, and the iPSCs may involve tumorigenicity caused by the integration of the transgenes. Recent studies have shown that somatic fibroblasts can be directly reprogrammed to NSCs, neurons, and specific dopamine neurons. Directly induced neurons (iN) or induced DA neurons (iDANs) from somatic fibroblasts have several advantages over iPSC cells. The neurons produced by direct transdifferentiation do not pass through a pluripotent state. Therefore, direct reprogramming can generate patient-specific cells, and it can overcome the safety problems of rejection by the immune system and teratoma formation related to hESCs and iPSCs. However, there are some critical issues such as the low efficiency of direct reprogramming, biological functions, and risks from the directly converted neurons, which hinder their clinical applications. Here, the recent progress in methods, mechanisms, and future challenges of directly reprogramming somatic fibroblasts into neurons or dopamine neurons were summarized to speed up the clinical translation of these directly converted neural cells to treat PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabin Han
- Innovation Institute for Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shenzhen Research Institute of Shandong University, Jinan, China.,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Yanming Liu
- Shenzhen Research Institute of Shandong University, Jinan, China.,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Jin Huang
- Laboratory of Basic Medical Research, Medical Centre of PLA Strategic Support Force, Beijing, China
| | - Xiaoping Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanfei Wei
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
24
|
Mertens J, Herdy JR, Traxler L, Schafer ST, Schlachetzki JCM, Böhnke L, Reid DA, Lee H, Zangwill D, Fernandes DP, Agarwal RK, Lucciola R, Zhou-Yang L, Karbacher L, Edenhofer F, Stern S, Horvath S, Paquola ACM, Glass CK, Yuan SH, Ku M, Szücs A, Goldstein LSB, Galasko D, Gage FH. Age-dependent instability of mature neuronal fate in induced neurons from Alzheimer's patients. Cell Stem Cell 2021; 28:1533-1548.e6. [PMID: 33910058 PMCID: PMC8423435 DOI: 10.1016/j.stem.2021.04.004] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/17/2021] [Accepted: 04/02/2021] [Indexed: 12/12/2022]
Abstract
Sporadic Alzheimer's disease (AD) exclusively affects elderly people. Using direct conversion of AD patient fibroblasts into induced neurons (iNs), we generated an age-equivalent neuronal model. AD patient-derived iNs exhibit strong neuronal transcriptome signatures characterized by downregulation of mature neuronal properties and upregulation of immature and progenitor-like signaling pathways. Mapping iNs to longitudinal neuronal differentiation trajectory data demonstrated that AD iNs reflect a hypo-mature neuronal identity characterized by markers of stress, cell cycle, and de-differentiation. Epigenetic landscape profiling revealed an underlying aberrant neuronal state that shares similarities with malignant transformation and age-dependent epigenetic erosion. To probe for the involvement of aging, we generated rejuvenated iPSC-derived neurons that showed no significant disease-related transcriptome signatures, a feature that is consistent with epigenetic clock and brain ontogenesis mapping, which indicate that fibroblast-derived iNs more closely reflect old adult brain stages. Our findings identify AD-related neuronal changes as age-dependent cellular programs that impair neuronal identity.
Collapse
Affiliation(s)
- Jerome Mertens
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria.
| | - Joseph R Herdy
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria; Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Larissa Traxler
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Simon T Schafer
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Lena Böhnke
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Dylan A Reid
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hyungjun Lee
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Dina Zangwill
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Diana P Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ravi K Agarwal
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Raffaella Lucciola
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lucia Zhou-Yang
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Lukas Karbacher
- Neural Aging Laboratory, Institute of Molecular Biology, CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Frank Edenhofer
- Department of Genomics, Stem Cells & Regenerative Medicine, Institute of Molecular Biology, CMBI, Institute of Molecular Biology and CMBI, Leopold-Franzens-University Innsbruck, Tyrol, Austria
| | - Shani Stern
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Steve Horvath
- Department of Human Genetics, Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Apua C M Paquola
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Shauna H Yuan
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; University of Minnesota, Twin Cities, Department of Neurology, Minneapolis, MN, USA
| | - Manching Ku
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Attila Szücs
- Neuronal Cell Biology Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Lawrence S B Goldstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
25
|
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]
|
26
|
Abstract
Progressive aging is a physiological process that represents a central risk factor for the development of several human age-associated chronic diseases, including neurodegenerative diseases. A major focus in biomedical research is the pursuit for appropriate model systems to better model the biology of human aging and the interface between aging and disease mechanisms. Direct conversion of human fibroblasts into induced neurons (iNs) has emerged as a novel technology for the in vitro modeling of age-dependent neurological diseases. Similar to other cellular reprogramming techniques, e.g., iPSC-based cellular reprograming, direct conversion relies on the ectopic overexpression of transcription factors, typically including well-known pioneer factors. However, in contrast to alternative technologies to generate neurons, the entire process of direct conversion bypasses any proliferative or stem cell-like stage, which in fact renders it the unique aptitude of preserving age-associated hallmarks from the initial fibroblast source. In this chapter, we introduce direct conversion as a practical and easy-to-approach disease model for aging and neurodegenerative disease research. A focus here is to provide a stepwise protocol for the efficient and highly reproducible generation of iNs from adult dermal fibroblasts from human donors.
Collapse
|
27
|
Kempf J, Knelles K, Hersbach BA, Petrik D, Riedemann T, Bednarova V, Janjic A, Simon-Ebert T, Enard W, Smialowski P, Götz M, Masserdotti G. Heterogeneity of neurons reprogrammed from spinal cord astrocytes by the proneural factors Ascl1 and Neurogenin2. Cell Rep 2021; 36:109409. [PMID: 34289357 PMCID: PMC8316252 DOI: 10.1016/j.celrep.2021.109409] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/14/2021] [Accepted: 06/24/2021] [Indexed: 01/21/2023] Open
Abstract
Astrocytes are a viable source for generating new neurons via direct conversion. However, little is known about the neurogenic cascades triggered in astrocytes from different regions of the CNS. Here, we examine the transcriptome induced by the proneural factors Ascl1 and Neurog2 in spinal cord-derived astrocytes in vitro. Each factor initially elicits different neurogenic programs that later converge to a V2 interneuron-like state. Intriguingly, patch sequencing (patch-seq) shows no overall correlation between functional properties and the transcriptome of the heterogenous induced neurons, except for K-channels. For example, some neurons with fully mature electrophysiological properties still express astrocyte genes, thus calling for careful molecular and functional analysis. Comparing the transcriptomes of spinal cord- and cerebral-cortex-derived astrocytes reveals profound differences, including developmental patterning cues maintained in vitro. These relate to the distinct neuronal identity elicited by Ascl1 and Neurog2 reflecting their developmental functions in subtype specification of the respective CNS region.
Collapse
Affiliation(s)
- J Kempf
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - K Knelles
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - B A Hersbach
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Graduate School of Systemic Neurosciences, LMU Munich, Planegg-Martinsried 82152, Germany
| | - D Petrik
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; School of Biosciences, The Sir Martin Evans Building, Cardiff University, CF10 3AX Cardiff, UK
| | - T Riedemann
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - V Bednarova
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - A Janjic
- Anthropology and Human Genomics, Faculty of Biology, LMU Munich, Planegg-Martinsried 82152, Germany
| | - T Simon-Ebert
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany
| | - W Enard
- Biomedical Center Munich, Bioinformatic Core Facility, LMU Munich, Planegg-Martinsried 82152, Germany
| | - P Smialowski
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; School of Biosciences, The Sir Martin Evans Building, Cardiff University, CF10 3AX Cardiff, UK
| | - M Götz
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany; Excellence Cluster of Systems Neurology (SYNERGY), Munich, Germany.
| | - G Masserdotti
- Biomedical Center Munich, Physiological Genomics, LMU Munich, Planegg-Martinsried 82152, Germany; Institute for Stem Cell Research, Helmholtz Center Munich, Neuherberg 85764, Germany.
| |
Collapse
|
28
|
Zhou M, Tao X, Sui M, Cui M, Liu D, Wang B, Wang T, Zheng Y, Luo J, Mu Y, Wan F, Zhu LQ, Zhang B. Reprogramming astrocytes to motor neurons by activation of endogenous Ngn2 and Isl1. Stem Cell Reports 2021; 16:1777-1791. [PMID: 34171285 PMCID: PMC8282467 DOI: 10.1016/j.stemcr.2021.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/13/2022] Open
Abstract
Central nervous system injury and neurodegenerative diseases cause irreversible loss of neurons. Overexpression of exogenous specific transcription factors can reprogram somatic cells into functional neurons for regeneration and functional reconstruction. However, these practices are potentially problematic due to the integration of vectors into the host genome. Here, we showed that the activation of endogenous genes Ngn2 and Isl1 by CRISPRa enabled reprogramming of mouse spinal astrocytes and embryonic fibroblasts to motor neurons. These induced neurons showed motor neuronal morphology and exhibited electrophysiological activities. Furthermore, astrocytes in the spinal cord of the adult mouse can be converted into motor neurons by this approach with high efficiency. These results demonstrate that the activation of endogenous genes is sufficient to induce astrocytes into functional motor neurons in vitro and in vivo. This direct neuronal reprogramming approach may provide a novel potential therapeutic strategy for treating neurodegenerative diseases and spinal cord injury.
Collapse
Affiliation(s)
- Meiling Zhou
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoqing Tao
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Ming Sui
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengge Cui
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Liu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Beibei Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yunjie Zheng
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Juan Luo
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yangling Mu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Feng Wan
- Department of Neurosurgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
29
|
One-step Reprogramming of Human Fibroblasts into Oligodendrocyte-like Cells by SOX10, OLIG2, and NKX6.2. Stem Cell Reports 2021; 16:771-783. [PMID: 33770499 PMCID: PMC8072064 DOI: 10.1016/j.stemcr.2021.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 01/31/2023] Open
Abstract
Limited access to human oligodendrocytes impairs better understanding of oligodendrocyte pathology in myelin diseases. Here, we describe a method to robustly convert human fibroblasts directly into oligodendrocyte-like cells (dc-hiOLs), which allows evaluation of remyelination-promoting compounds and disease modeling. Ectopic expression of SOX10, OLIG2, and NKX6.2 in human fibroblasts results in rapid generation of O4+ cells, which further differentiate into MBP+ mature oligodendrocyte-like cells within 16 days. dc-hiOLs undergo chromatin remodeling to express oligodendrocyte markers, ensheath axons, and nanofibers in vitro, respond to promyelination compound treatment, and recapitulate in vitro oligodendroglial pathologies associated with Pelizaeus-Merzbacher leukodystrophy related to PLP1 mutations. Furthermore, DNA methylome analysis provides evidence that the CpG methylation pattern significantly differs between dc-hiOLs derived from fibroblasts of young and old donors, indicating the maintenance of the source cells’ “age.” In summary, dc-hiOLs represent a reproducible technology that could contribute to personalized medicine in the field of myelin diseases. SOX10, OLIG2, and NKX6.2 directly convert human fibroblasts into dc-hiOLs in 16 days dc-hiOLs express key oligodendrocyte markers dc-hiOLs preserve the epigenetic age of donor cells dc-hiOLs from PMD patients show maturation deficit and vulnerability to cell death
Collapse
|
30
|
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
|
31
|
Cates K, McCoy MJ, Kwon JS, Liu Y, Abernathy DG, Zhang B, Liu S, Gontarz P, Kim WK, Chen S, Kong W, Ho JN, Burbach KF, Gabel HW, Morris SA, Yoo AS. Deconstructing Stepwise Fate Conversion of Human Fibroblasts to Neurons by MicroRNAs. Cell Stem Cell 2021; 28:127-140.e9. [PMID: 32961143 PMCID: PMC7796891 DOI: 10.1016/j.stem.2020.08.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 07/11/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022]
Abstract
Cell-fate conversion generally requires reprogramming effectors to both introduce fate programs of the target cell type and erase the identity of starting cell population. Here, we reveal insights into the activity of microRNAs miR-9/9∗ and miR-124 (miR-9/9∗-124) as reprogramming agents that orchestrate direct conversion of human fibroblasts into motor neurons by first eradicating fibroblast identity and promoting uniform transition to a neuronal state in sequence. We identify KLF-family transcription factors as direct target genes for miR-9/9∗-124 and show their repression is critical for erasing fibroblast fate. Subsequent gain of neuronal identity requires upregulation of a small nuclear RNA, RN7SK, which induces accessibilities of chromatin regions and neuronal gene activation to push cells to a neuronal state. Our study defines deterministic components in the microRNA-mediated reprogramming cascade.
Collapse
Affiliation(s)
- Kitra Cates
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J McCoy
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ji-Sun Kwon
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Computational and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yangjian Liu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel G Abernathy
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Developmental, Regenerative, and Stem Cell Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shaopeng Liu
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul Gontarz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Woo Kyung Kim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shawei Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenjun Kong
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Computational and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua N Ho
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Developmental, Regenerative, and Stem Cell Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyle F Burbach
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Program in Molecular Genetics and Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harrison W Gabel
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samantha A Morris
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew S Yoo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
32
|
Dotson GA, Ryan CW, Chen C, Muir L, Rajapakse I. Cellular reprogramming: Mathematics meets medicine. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 13:e1515. [PMID: 33289324 PMCID: PMC8867497 DOI: 10.1002/wsbm.1515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 11/11/2022]
Abstract
Generating needed cell types using cellular reprogramming is a promising strategy for restoring tissue function in injury or disease. A common method for reprogramming is addition of one or more transcription factors that confer a new function or identity. Advancements in transcription factor selection and delivery have culminated in successful grafting of autologous reprogrammed cells, an early demonstration of their clinical utility. Though cellular reprogramming has been successful in a number of settings, identification of appropriate transcription factors for a particular transformation has been challenging. Computational methods enable more sophisticated prediction of relevant transcription factors for reprogramming by leveraging gene expression data of initial and target cell types, and are built on mathematical frameworks ranging from information theory to control theory. This review highlights the utility and impact of these mathematical frameworks in the field of cellular reprogramming. This article is categorized under: Reproductive System Diseases > Reproductive System Diseases>Genetics/Genomics/Epigenetics Reproductive System Diseases > Reproductive System Diseases>Stem Cells and Development Reproductive System Diseases > Reproductive System Diseases>Computational Models.
Collapse
Affiliation(s)
- Gabrielle A. Dotson
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Charles W. Ryan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Can Chen
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Lindsey Muir
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Indika Rajapakse
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, 48109, USA
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| |
Collapse
|
33
|
Ma S, Zang T, Liu ML, Zhang CL. Aging-relevant human basal forebrain cholinergic neurons as a cell model for Alzheimer's disease. Mol Neurodegener 2020; 15:61. [PMID: 33087140 PMCID: PMC7579825 DOI: 10.1186/s13024-020-00411-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Background Alzheimer’s disease (AD) is an adult-onset mental disorder with aging as a major risk factor. Early and progressive degeneration of basal forebrain cholinergic neurons (BFCNs) contributes substantially to cognitive impairments of AD. An aging-relevant cell model of BFCNs will critically help understand AD and identify potential therapeutics. Recent studies demonstrate that induced neurons directly reprogrammed from adult human skin fibroblasts retain aging-associated features. However, human induced BFCNs (hiBFCNs) have yet to be achieved. Methods We examined a reprogramming procedure for the generation of aging-relevant hiBFCNs through virus-mediated expression of fate-determining transcription factors. Skin fibroblasts were obtained from healthy young persons, healthy adults and sporadic AD patients. Properties of the induced neurons were examined by immunocytochemistry, qRT-PCR, western blotting, and electrophysiology. Results We established a protocol for efficient generation of hiBFCNs from adult human skin fibroblasts. They show electrophysiological properties of mature neurons and express BFCN-specific markers, such as CHAT, p75NTR, ISL1, and VACHT. As a proof-of-concept, our preliminary results further reveal that hiBFCNs from sporadic AD patients exhibit time-dependent TAU hyperphosphorylation in the soma and dysfunctional nucleocytoplasmic transport activities. Conclusions Aging-relevant BFCNs can be directly reprogrammed from human skin fibroblasts of healthy adults and sporadic AD patients. They show promises as an aging-relevant cell model for understanding AD pathology and may be employed for therapeutics identification for AD.
Collapse
Affiliation(s)
- Shuaipeng Ma
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Tong Zang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Meng-Lu Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA. .,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| |
Collapse
|
34
|
Carter JL, Halmai JANM, Fink KD. The iNs and Outs of Direct Reprogramming to Induced Neurons. Front Genome Ed 2020; 2:7. [PMID: 34713216 PMCID: PMC8525349 DOI: 10.3389/fgeed.2020.00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Understanding of cell-type specific transcription factors has promoted progress in methods for cellular reprogramming, such as directly reprogramming somatic cells to induced neurons (iN). Methods for direct reprogramming require neuronal-fate determining gene activation via neuron-specific microRNAs, chemical modulation of key neuronal signaling pathways or overexpression via viral vectors, with some reprogramming strategies requiring a combination of these methods to induce the neuronal-cell fate. These methods have been employed in a multitude of cell types, including fibroblasts, hepatocytes, peripheral blood mononuclear, and T cells. The ability to create iN from skin biopsies and blood samples coupled with recent advancements in artificially inducing age- and disease-associated phenotypes are accelerating the development of disease models for late-onset neurodegenerative disorders. Here, we review how activation of the neuronal transcriptome alters the epigenetic landscape of the donor cell to facilitate reprogramming to neurons. We also discuss the advantages of using DNA binding domains such as CRISPR/dCas9 to overcome epigenetic barriers to induce neuronal-cell fate by activating endogenous neuronal cell-fate determining genes.
Collapse
|
35
|
A developed serum-free medium and an optimized chemical cocktail for direct conversion of human dermal fibroblasts into brown adipocytes. Sci Rep 2020; 10:3775. [PMID: 32111895 PMCID: PMC7048747 DOI: 10.1038/s41598-020-60769-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/17/2020] [Indexed: 01/12/2023] Open
Abstract
Brown adipocytes coordinate systemic energy metabolism associated with the pathogenesis of obesity and related metabolic diseases including type 2 diabetes. We have previously reported chemical compound-induced brown adipocytes (ciBAs) converted from human dermal fibroblasts without using transgenes. In this study, to reveal a precise molecular mechanism underlying the direct conversion and human adipocyte browning, we developed serum-free brown adipogenic medium (SFBAM) with an optimized chemical cocktail consisting of Rosiglitazone, Forskolin, and BMP7. During the direct conversion, treatment with BMP7 enhanced Ucp1 expression rather than the conversion efficiency in the absence of BMP signalling inhibitors. Moreover, treatment with a TGF-β signalling pathway inhibitor was no longer required in the serum-free medium, likely because the TGF-β pathway was already suppressed. SFBAM and the chemical cocktail efficiently converted human dermal fibroblasts into ciBAs within four weeks. The ciBAs exhibited increased mitochondrial levels, elevated oxygen consumption rate, and a response to β-adrenergic receptor agonists. Thus the ciBAs converted by the serum-free medium and the chemical cocktail provide a novel model of human brown (beige) adipocytes applicable for basic research, drug screening, and clinical applications.
Collapse
|
36
|
Schlachetzki JCM, Toda T, Mertens J. When function follows form: Nuclear compartment structure and the epigenetic landscape of the aging neuron. Exp Gerontol 2020; 133:110876. [PMID: 32068088 DOI: 10.1016/j.exger.2020.110876] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 12/21/2022]
Abstract
The human brain is affected by cellular aging. Neurons are primarily generated during embryogenesis and early life with a limited capacity for renewal and replacement, making them some of the oldest cells in the human body. Our present understanding of neurodegenerative diseases points towards advanced neuronal age as a prerequisite for the development of these disorders. While significant progress has been made in understanding the relationship between aging and neurological disease, it will be essential to delve further into the molecular mechanisms of neuronal aging in order to develop therapeutic interventions targeting age-related brain dysfunction. In this mini review, we highlight recent findings on the relationship between the aging of nuclear structures and changes in the epigenetic landscape during neuronal aging and disease.
Collapse
Affiliation(s)
- Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tomohisa Toda
- Nuclear Architecture in Neural Plasticity and Aging, German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.
| | - Jerome Mertens
- Institute of Molecular Biology & CMBI, Leopold-Franzens-University Innsbruck, Austria; Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
37
|
HORISAWA K, SUZUKI A. Direct cell-fate conversion of somatic cells: Toward regenerative medicine and industries. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2020; 96:131-158. [PMID: 32281550 PMCID: PMC7247973 DOI: 10.2183/pjab.96.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Cells of multicellular organisms have diverse characteristics despite having the same genetic identity. The distinctive phenotype of each cell is determined by molecular mechanisms such as epigenetic changes that occur throughout the lifetime of an individual. Recently, technologies that enable modification of the fate of somatic cells have been developed, and the number of studies using these technologies has increased drastically in the last decade. Various cell types, including neuronal cells, cardiomyocytes, and hepatocytes, have been generated using these technologies. Although most direct reprogramming methods employ forced transduction of a defined sets of transcription factors to reprogram cells in a manner similar to induced pluripotent cell technology, many other strategies, such as methods utilizing chemical compounds and microRNAs to change the fate of somatic cells, have also been developed. In this review, we summarize transcription factor-based reprogramming and various other reprogramming methods. Additionally, we describe the various industrial applications of direct reprogramming technologies.
Collapse
Affiliation(s)
- Kenichi HORISAWA
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi SUZUKI
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
- Correspondence should be addressed: A. Suzuki, Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan (e-mail: )
| |
Collapse
|
38
|
Traxler L, Edenhofer F, Mertens J. Next-generation disease modeling with direct conversion: a new path to old neurons. FEBS Lett 2019; 593:3316-3337. [PMID: 31715002 PMCID: PMC6907729 DOI: 10.1002/1873-3468.13678] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/20/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022]
Abstract
Within just over a decade, human reprogramming-based disease modeling has developed from a rather outlandish idea into an essential part of disease research. While iPSCs are a valuable tool for modeling developmental and monogenetic disorders, their rejuvenated identity poses limitations for modeling age-associated diseases. Direct cell-type conversion of fibroblasts into induced neurons (iNs) circumvents rejuvenation and preserves hallmarks of cellular aging. iNs are thus advantageous for modeling diseases that possess strong age-related and epigenetic contributions and can complement iPSC-based strategies for disease modeling. In this review, we provide an overview of the state of the art of direct iN conversion and describe the key epigenetic, transcriptomic, and metabolic changes that occur in converting fibroblasts. Furthermore, we summarize new insights into this fascinating process, particularly focusing on the rapidly changing criteria used to define and characterize in vitro-born human neurons. Finally, we discuss the unique features that distinguish iNs from other reprogramming-based neuronal cell models and how iNs are relevant to disease modeling.
Collapse
Affiliation(s)
- Larissa Traxler
- Department of GenomicsStem Cell Biology & Regenerative MedicineInstitute of Molecular Biology & CMBILeopold‐Franzens‐University InnsbruckInnsbruckAustria
- Laboratory of GeneticsThe Salk Institute for Biological StudiesLa JollaCAUSA
| | - Frank Edenhofer
- Department of GenomicsStem Cell Biology & Regenerative MedicineInstitute of Molecular Biology & CMBILeopold‐Franzens‐University InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Department of GenomicsStem Cell Biology & Regenerative MedicineInstitute of Molecular Biology & CMBILeopold‐Franzens‐University InnsbruckInnsbruckAustria
- Laboratory of GeneticsThe Salk Institute for Biological StudiesLa JollaCAUSA
| |
Collapse
|
39
|
Chen S, Zhang J, Zhang D, Jiao J. Acquisition of functional neurons by direct conversion: Switching the developmental clock directly. J Genet Genomics 2019; 46:459-465. [PMID: 31771824 DOI: 10.1016/j.jgg.2019.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/20/2019] [Accepted: 10/08/2019] [Indexed: 01/04/2023]
Abstract
Identifying approaches for treating neurodegeneration is a thorny task but is important for a growing number of patients. Researchers have focused on discovering the underlying molecular mechanisms of reprogramming and optimizing the technologies for acquiring neurons. Direct conversion is one of the most important processes for treating neurological disorders. Induced neurons derived from direct conversion, which bypass the pluripotency stage, are more effective, more quickly obtained, and are safer than those produced via induced pluripotent stem cells (iPSCs). Based on iPSC strategies, scientists have derived methods to obtain functional neurons by direct conversion, such as neuron-related transcriptional factors, small molecules, microRNAs, and epigenetic modifiers. In this review, we discuss the present strategies for direct conversion of somatic cells into functional neurons and the potentials of direct conversion for producing functional neurons and treating neurodegeneration.
Collapse
Affiliation(s)
- Shuangquan Chen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Juan Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Dongming Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwei Jiao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
40
|
Pereira M, Birtele M, Rylander Ottosson D. Direct reprogramming into interneurons: potential for brain repair. Cell Mol Life Sci 2019; 76:3953-3967. [PMID: 31250034 PMCID: PMC6785593 DOI: 10.1007/s00018-019-03193-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/29/2022]
Abstract
The brain tissue has only a limited capacity for generating new neurons. Therefore, to treat neurological diseases, there is a need of other cell sources for brain repair. Different sources of cells have been subject of intense research over the years, including cells from primary tissue, stem cell-derived cells and reprogrammed cells. As an alternative, direct reprogramming of resident brain cells into neurons is a recent approach that could provide an attractive method for treating brain injuries or diseases as it uses the patient's own cells for generating novel neurons inside the brain. In vivo reprogramming is still in its early stages but holds great promise as an option for cell therapy. To date, both inhibitory and excitatory neurons have been obtained via in vivo reprogramming, but the precise phenotype or functionality of these cells has not been analysed in detail in most of the studies. Recent data shows that in vivo reprogrammed neurons are able to functionally mature and integrate into the existing brain circuitry, and compose interneuron phenotypes that seem to correlate to their endogenous counterparts. Interneurons are of particular importance as they are essential in physiological brain function and when disturbed lead to several neurological disorders. In this review, we describe a comprehensive overview of the existing studies involving brain repair, including in vivo reprogramming, with a focus on interneurons, along with an overview on current efforts to generate interneurons for cell therapy for a number of neurological diseases.
Collapse
Affiliation(s)
- Maria Pereira
- Department of Experimental Medical Science and Lund Stem Cell Center BMC, Lund University, 22141, Lund, Sweden
| | - Marcella Birtele
- Department of Experimental Medical Science and Lund Stem Cell Center BMC, Lund University, 22141, Lund, Sweden
| | - Daniella Rylander Ottosson
- Department of Experimental Medical Science and Lund Stem Cell Center BMC, Lund University, 22141, Lund, Sweden.
| |
Collapse
|
41
|
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
|