Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      4D Genome Rewiring during Oncogene-Induced and Replicative Senescence

      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.

          Summary

          To understand the role of the extensive senescence-associated 3D genome reorganization, we generated genome-wide chromatin interaction maps, epigenome, replication-timing, whole-genome bisulfite sequencing, and gene expression profiles from cells entering replicative senescence (RS) or upon oncogene-induced senescence (OIS). We identify senescence-associated heterochromatin domains (SAHDs). Differential intra- versus inter-SAHD interactions lead to the formation of senescence-associated heterochromatin foci (SAHFs) in OIS but not in RS. This OIS-specific configuration brings active genes located in genomic regions adjacent to SAHDs in close spatial proximity and favors their expression. We also identify DNMT1 as a factor that induces SAHFs by promoting HMGA2 expression. Upon DNMT1 depletion, OIS cells transition to a 3D genome conformation akin to that of cells in replicative senescence. These data show how multi-omics and imaging can identify critical features of RS and OIS and discover determinants of acute senescence and SAHF formation.

          Graphical Abstract

          Highlights

          • Deep multi-omics characterization of replicative and oncogene-induced senescence

          • Senescence-associated heterochromatin domains (SAHDs) form SAHFs via 3D changes

          • DNMT1 is required for SAHF formation via regulation of HMGA2 expression

          • SAHF formation leads to expression of SAHF-adjacent genes via 3D chromatin contacts

          Abstract

          Sati et al. studied 3D chromatin organization in different types of cellular senescence. They have identified DNMT1 and HMGA2-mediated changes in the structural organization of senescence-associated heterochromatin domains (SAHDs) and architecture-associated gene-expression changes as the key difference among different senescent systems.

          Related collections

          Most cited references41

          • Record: found
          • Abstract: found
          • Article: found

          Aging, Cellular Senescence, and Cancer

          For most species, aging promotes a host of degenerative pathologies that are characterized by debilitating losses of tissue or cellular function. However, especially among vertebrates, aging also promotes hyperplastic pathologies, the most deadly of which is cancer. In contrast to the loss of function that characterizes degenerating cells and tissues, malignant (cancerous) cells must acquire new (albeit aberrant) functions that allow them to develop into a lethal tumor. This review discusses the idea that, despite seemingly opposite characteristics, the degenerative and hyperplastic pathologies of aging are at least partly linked by a common biological phenomenon: a cellular stress response known as cellular senescence. The senescence response is widely recognized as a potent tumor suppressive mechanism. However, recent evidence strengthens the idea that it also drives both degenerative and hyperplastic pathologies, most likely by promoting chronic inflammation. Thus, the senescence response may be the result of antagonistically pleiotropic gene action.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a.

            Oncogenic ras can transform most immortal rodent cells to a tumorigenic state. However, transformation of primary cells by ras requires either a cooperating oncogene or the inactivation of tumor suppressors such as p53 or p16. Here we show that expression of oncogenic ras in primary human or rodent cells results in a permanent G1 arrest. The arrest induced by ras is accompanied by accumulation of p53 and p16, and is phenotypically indistinguishable from cellular senescence. Inactivation of either p53 or p16 prevents ras-induced arrest in rodent cells, and E1A achieves a similar effect in human cells. These observations suggest that the onset of cellular senescence does not simply reflect the accumulation of cell divisions, but can be prematurely activated in response to an oncogenic stimulus. Negation of ras-induced senescence may be relevant during multistep tumorigenesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state.

              Direct reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) provides a unique opportunity to derive patient-specific stem cells with potential applications in tissue replacement therapies and without the ethical concerns of human embryonic stem cells (hESCs). However, cellular senescence, which contributes to aging and restricted longevity, has been described as a barrier to the derivation of iPSCs. Here we demonstrate, using an optimized protocol, that cellular senescence is not a limit to reprogramming and that age-related cellular physiology is reversible. Thus, we show that our iPSCs generated from senescent and centenarian cells have reset telomere size, gene expression profiles, oxidative stress, and mitochondrial metabolism, and are indistinguishable from hESCs. Finally, we show that senescent and centenarian-derived pluripotent stem cells are able to redifferentiate into fully rejuvenated cells. These results provide new insights into iPSC technology and pave the way for regenerative medicine for aged patients.
                Bookmark

                Author and article information

                Contributors
                Journal
                Mol Cell
                Mol. Cell
                Molecular Cell
                Cell Press
                1097-2765
                1097-4164
                07 May 2020
                07 May 2020
                : 78
                : 3
                : 522-538.e9
                Affiliations
                [1 ]Institute of Human Genetics, UMR 9002, CNRS and University of Montpellier, Montpellier, France
                [2 ]Institute for Regenerative Medicine and Biotherapy, Univ Montpellier, INSERM UMR1183, F-34295 Montpellier, France
                [3 ]Univ Grenoble Alpes, CNRS, Grenoble INP, TIMC-IMAG, 38000 Grenoble, France
                [4 ]CNAG-CRG, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
                [5 ]Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
                [6 ]Department of Biological Science and Center for Genomics and Personalized Medicine, Florida State University, Tallahassee, FL 32306, USA
                [7 ]Unit of Chromosomal Genetics and Chromostem Research Platform, CHU, Montpellier, France
                [8 ]Barcelona Supercomputing Center, Barcelona, Spain
                [9 ]Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
                [10 ]Institute for Research in Biomedicine, the Barcelona Institute of Science and Technology, Barcelona, Spain
                [11 ]Laboratoire de Physique (UMR CNRS 5672), ENS de Lyon, Lyon, France
                [12 ]Centre for Genomic Regulation, The Barcelona Institute for Science and Technology, Carrer del Doctor Aiguader 88, Barcelona 08003, Spain
                [13 ]Pompeu Fabra University, Doctor Aiguader 88, Barcelona 08003, Spain
                [14 ]CREA, PgLluís Companys 23, 08010 Barcelona, Spain
                [15 ]CHRU de Montpellier, Montpellier, France
                Author notes
                []Corresponding author jean-marc.lemaitre@ 123456inserm.fr
                [∗∗ ]Corresponding author giacomo.cavalli@ 123456igh.cnrs.fr
                [16]

                Present address: Pioneer Campus, Helmholz Zentrum München, Neuherberg, Germany

                [17]

                Present address: Université Lyon, ENS de Lyon, Université Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 69007 Lyon, France

                [18]

                These authors contributed equally

                [19]

                Lead Contact

                Article
                S1097-2765(20)30155-6
                10.1016/j.molcel.2020.03.007
                7208559
                32220303
                456ed9a1-c4a8-423b-9f2b-b65acb23c895
                © 2020 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 16 May 2019
                : 20 December 2019
                : 4 March 2020
                Categories
                Article

                Molecular biology
                senescence,replicative senescence,oncogene-induced senescence,3d genome architecture,hi-c,chromatin compartments,gene regulation,dnmt1

                Comments

                Comment on this article