9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Next‐generation disease modeling with direct conversion: a new path to old neurons

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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.

          Abstract

          Related collections

          Most cited references124

          • Record: found
          • Abstract: found
          • Article: not found

          Direct conversion of fibroblasts to functional neurons by defined factors

          Cellular differentiation and lineage commitment are considered robust and irreversible processes during development. Recent work has shown that mouse and human fibroblasts can be reprogrammed to a pluripotent state with a combination of four transcription factors. This raised the question of whether transcription factors could directly induce other defined somatic cell fates, and not only an undifferentiated state. We hypothesized that combinatorial expression of neural lineage-specific transcription factors could directly convert fibroblasts into neurons. Starting from a pool of nineteen candidate genes, we identified a combination of only three factors, Ascl1, Brn2, and Myt1l, that suffice to rapidly and efficiently convert mouse embryonic and postnatal fibroblasts into functional neurons in vitro. These induced neuronal (iN) cells express multiple neuron-specific proteins, generate action potentials, and form functional synapses. Generation of iN cells from non-neural lineages could have important implications for studies of neural development, neurological disease modeling, and regenerative medicine.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            An epigenetic biomarker of aging for lifespan and healthspan

            Identifying reliable biomarkers of aging is a major goal in geroscience. While the first generation of epigenetic biomarkers of aging were developed using chronological age as a surrogate for biological age, we hypothesized that incorporation of composite clinical measures of phenotypic age that capture differences in lifespan and healthspan may identify novel CpGs and facilitate the development of a more powerful epigenetic biomarker of aging. Using an innovative two-step process, we develop a new epigenetic biomarker of aging, DNAm PhenoAge, that strongly outperforms previous measures in regards to predictions for a variety of aging outcomes, including all-cause mortality, cancers, healthspan, physical functioning, and Alzheimer's disease. While this biomarker was developed using data from whole blood, it correlates strongly with age in every tissue and cell tested. Based on an in-depth transcriptional analysis in sorted cells, we find that increased epigenetic, relative to chronological age, is associated with increased activation of pro-inflammatory and interferon pathways, and decreased activation of transcriptional/translational machinery, DNA damage response, and mitochondrial signatures. Overall, this single epigenetic biomarker of aging is able to capture risks for an array of diverse outcomes across multiple tissues and cells, and provide insight into important pathways in aging.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation.

              The energy requirements of the brain are very high, and tight regulatory mechanisms operate to ensure adequate spatial and temporal delivery of energy substrates in register with neuronal activity. Astrocytes-a type of glial cell-have emerged as active players in brain energy delivery, production, utilization, and storage. Our understanding of neuroenergetics is rapidly evolving from a "neurocentric" view to a more integrated picture involving an intense cooperativity between astrocytes and neurons. This review focuses on the cellular aspects of brain energy metabolism, with a particular emphasis on the metabolic interactions between neurons and astrocytes. Copyright © 2011 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                jerome.mertens@uibk.ac.at
                Journal
                FEBS Lett
                FEBS Lett
                10.1002/(ISSN)1873-3468
                FEB2
                Febs Letters
                John Wiley and Sons Inc. (Hoboken )
                0014-5793
                1873-3468
                26 November 2019
                December 2019
                : 593
                : 23 , Neural and Hematopoietic Stem Cell Reprogramming ( doiID: 10.1002/feb2.v593.23 )
                : 3316-3337
                Affiliations
                [ 1 ] Department of Genomics Stem Cell Biology & Regenerative Medicine Institute of Molecular Biology & CMBI Leopold‐Franzens‐University Innsbruck Innsbruck Austria
                [ 2 ] Laboratory of Genetics The Salk Institute for Biological Studies La Jolla CA USA
                Author notes
                [*] [* ] Correspondence

                J. Mertens, Department of Genomics, Stem Cell Biology & Regenerative Medicine, Institute of Molecular Biology & CMBI, University of Innsbruck, Technikerstr. 25, 6020 Innsbruck, Austria

                Tel: +43 512 507 51413

                E‐mail: jerome.mertens@ 123456uibk.ac.at

                Author information
                https://orcid.org/0000-0003-0974-3575
                https://orcid.org/0000-0002-6489-714X
                https://orcid.org/0000-0002-4291-4121
                Article
                FEB213678
                10.1002/1873-3468.13678
                6907729
                31715002
                d89c5e41-24cb-45b7-ade0-f238e8b53fc6
                © 2019 The Authors. FEBS Letters published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 20 August 2019
                : 20 October 2019
                : 07 November 2019
                Page count
                Figures: 2, Tables: 1, Pages: 22, Words: 16215
                Funding
                Funded by: National Institutes of Health Pathway to Independence Award
                Award ID: K99‐AG056679‐01
                Funded by: European Union's Horizon 2020 research and innovation program
                Award ID: H2020‐MSCA‐IF‐2017 797205
                Funded by: Austrian Science Fund , open-funder-registry 10.13039/501100002428;
                Award ID: DK‐W1206
                Funded by: BrightFocus Foundation , open-funder-registry 10.13039/100006312;
                Award ID: A2019562S
                Categories
                Review Article
                Neuroscience
                Custom metadata
                2.0
                December 2019
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.7.3 mode:remove_FC converted:17.12.2019

                Molecular biology
                aging,cellular reprogramming,direct conversion,disease modeling,epigenetics,geriatric diseases,induced neurons,metabolism,neurodegenerative disorders

                Comments

                Comment on this article