17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      REST suppression mediates neural conversion of adult human fibroblasts via microRNA‐dependent and ‐independent pathways

      research-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

          Direct conversion of human fibroblasts into mature and functional neurons, termed induced neurons ( iNs), was achieved for the first time 6 years ago. This technology offers a promising shortcut for obtaining patient‐ and disease‐specific neurons for disease modeling, drug screening, and other biomedical applications. However, fibroblasts from adult donors do not reprogram as easily as fetal donors, and no current reprogramming approach is sufficiently efficient to allow the use of this technology using patient‐derived material for large‐scale applications. Here, we investigate the difference in reprogramming requirements between fetal and adult human fibroblasts and identify REST as a major reprogramming barrier in adult fibroblasts. Via functional experiments where we overexpress and knockdown the REST‐controlled neuron‐specific micro RNAs miR‐9 and miR‐124, we show that the effect of REST inhibition is only partially mediated via micro RNA up‐regulation. Transcriptional analysis confirmed that REST knockdown activates an overlapping subset of neuronal genes as micro RNA overexpression and also a distinct set of neuronal genes that are not activated via micro RNA overexpression. Based on this, we developed an optimized one‐step method to efficiently reprogram dermal fibroblasts from elderly individuals using a single‐vector system and demonstrate that it is possible to obtain iNs of high yield and purity from aged individuals with a range of familial and sporadic neurodegenerative disorders including Parkinson's, Huntington's, as well as Alzheimer's disease.

          Related collections

          Most cited references22

          • 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: not found

            Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution.

            Ectopic expression of the four transcription factors Oct4, Sox2, c-Myc, and Klf4 is sufficient to confer a pluripotent state upon the fibroblast genome, generating induced pluripotent stem (iPS) cells. It remains unknown if nuclear reprogramming induced by these four factors globally resets epigenetic differences between differentiated and pluripotent cells. Here, using novel selection approaches, we have generated iPS cells from fibroblasts to characterize their epigenetic state. Female iPS cells showed reactivation of a somatically silenced X chromosome and underwent random X inactivation upon differentiation. Genome-wide analysis of two key histone modifications indicated that iPS cells are highly similar to ES cells. Consistent with these observations, iPS cells gave rise to viable high-degree chimeras with contribution to the germline. These data show that transcription factor-induced reprogramming leads to the global reversion of the somatic epigenome into an ES-like state. Our results provide a paradigm for studying the epigenetic modifications that accompany nuclear reprogramming and suggest that abnormal epigenetic reprogramming does not pose a problem for the potential therapeutic applications of iPS cells.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Induced pluripotent stem cells from a spinal muscular atrophy patient.

              Spinal muscular atrophy is one of the most common inherited forms of neurological disease leading to infant mortality. Patients have selective loss of lower motor neurons resulting in muscle weakness, paralysis and often death. Although patient fibroblasts have been used extensively to study spinal muscular atrophy, motor neurons have a unique anatomy and physiology which may underlie their vulnerability to the disease process. Here we report the generation of induced pluripotent stem cells from skin fibroblast samples taken from a child with spinal muscular atrophy. These cells expanded robustly in culture, maintained the disease genotype and generated motor neurons that showed selective deficits compared to those derived from the child's unaffected mother. This is the first study to show that human induced pluripotent stem cells can be used to model the specific pathology seen in a genetically inherited disease. As such, it represents a promising resource to study disease mechanisms, screen new drug compounds and develop new therapies.
                Bookmark

                Author and article information

                Contributors
                malin.parmar@med.lu.se
                Journal
                EMBO Mol Med
                EMBO Mol Med
                10.1002/(ISSN)1757-4684
                EMMM
                embomm
                EMBO Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1757-4676
                1757-4684
                23 June 2017
                August 2017
                : 9
                : 8 ( doiID: 10.1002/emmm.v9.8 )
                : 1117-1131
                Affiliations
                [ 1 ] Division of Neurobiology and Lund Stem Cell Center Department of Experimental Medical Science Wallenberg Neuroscience Center Lund University Lund Sweden
                [ 2 ] John van Geest Centre for Brain Repair & Department of Neurology Department of Clinical Neurosciences University of Cambridge, Forvie Site Cambridge UK
                [ 3 ] Department of Experimental Medical Science Unit of Lung Biology BMC C12 Lund University Lund Sweden
                [ 4 ] Division for Neurogeriatrics Department of NVS Center for Alzheimer Research Karolinska Institutet Huddinge Sweden
                [ 5 ] Department of Geriatric Medicine Karolinska University Hospital Stockholm Sweden
                [ 6 ] Clinical Memory Research Unit Department of Clinical Sciences Malmö Lund University Lund Sweden
                Author notes
                [*] [* ]Corresponding author. Tel: +46 46 222 06 20; E‐mail: malin.parmar@ 123456med.lu.se
                [†]

                These authors contributed equally to this work

                Author information
                http://orcid.org/0000-0003-0669-7673
                http://orcid.org/0000-0001-5002-4199
                Article
                EMMM201607471
                10.15252/emmm.201607471
                5538296
                28646119
                edbce70a-d559-4f50-9472-1cc3b33b4c27
                © 2017 The Authors. Published under the terms of the CC BY 4.0 license

                This is an open access article under the terms of the Creative Commons Attribution 4.0 License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 16 December 2016
                : 12 May 2017
                : 18 May 2017
                Page count
                Figures: 6, Tables: 0, Pages: 15, Words: 10021
                Funding
                Funded by: New York Stem Cell Foundation (NYSCF)
                Funded by: H2020 | H2020 Priority Excellent Science | H2020 European Research Council (ERC)
                Award ID: 602278
                Award ID: 30971
                Award ID: FP/2007‐2013
                Funded by: Vetenskapsrådet (VR)
                Award ID: 521‐2012‐5624
                Award ID: 2016‐00873
                Award ID: 70862601
                Funded by: Swedish Parkinson's Disease Association | Parkinsonfonden (Parkinson Foundation)
                Award ID: 874/16
                Funded by: Gouvernement du Canada | Canadian Institutes of Health Research (CIHR)
                Award ID: 358492
                Funded by: Strategic Research Area at Lund University Multipark
                Funded by: Swedish Foundation for Strategic Research
                Award ID: FFL12‐0074
                Funded by: NIHR Biomedical Research Centre
                Funded by: University of Cambridge/Addenbrooke's Hospital
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                emmm201607471
                August 2017
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.1.4 mode:remove_FC converted:01.08.2017

                Molecular medicine
                adult human dermal fibroblasts,induced neurons,micrornas 9/9* and 124,re1‐silencing transcription factor,chromatin, epigenetics, genomics & functional genomics,neuroscience,stem cells

                Comments

                Comment on this article