Early growth response 2 (EGR2) is a novel regulator of the senescence programme

Senescence, a state of stable growth arrest, plays an important role in ageing and age‐related diseases in vivo. Although the INK4/ARF locus is known to be essential for senescence programmes, the key regulators driving p16 and ARF transcription remain largely underexplored. Using siRNA screening for modulators of the p16/pRB and ARF/p53/p21 pathways in deeply senescent human mammary epithelial cells (DS HMECs) and fibroblasts (DS HMFs), we identified EGR2 as a novel regulator of senescence. EGR2 expression is up‐regulated during senescence, and its ablation by siRNA in DS HMECs and HMFs transiently reverses the senescent phenotype. We demonstrate that EGR2 activates the ARF and p16 promoters and directly binds to both the ARF and p16 promoters. Loss of EGR2 down‐regulates p16 levels and increases the pool of p16− p21− ‘reversed’ cells in the population. Moreover, EGR2 overexpression is sufficient to induce senescence. Our data suggest that EGR2 is a direct transcriptional activator of the p16/pRB and ARF/p53/p21 pathways in senescence and a novel marker of senescence.

[1]  J. Cintrat,et al.  Glucocorticoids delay RAF-induced senescence promoted by EGR1 , 2019, Journal of Cell Science.

[2]  O. Bischof,et al.  AP-1 Imprints a Reversible Transcriptional Program of Senescent Cells , 2019, bioRxiv.

[3]  V. Brunton,et al.  The innate immune sensor Toll-like receptor 2 controls the senescence-associated secretory phenotype , 2018, Science Advances.

[4]  A. Pezeshki,et al.  Naturally occurring p16Ink4a-positive cells shorten healthy lifespan , 2016, Nature.

[5]  David J. Arenillas,et al.  JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles , 2015, Nucleic Acids Res..

[6]  Andres Metspalu,et al.  The transcriptional landscape of age in human peripheral blood , 2015, Nature Communications.

[7]  Robert Lowe,et al.  The senescent methylome and its relationship with cancer, ageing and germline genetic variation in humans , 2015, Genome Biology.

[8]  N. Sharpless,et al.  Forging a signature of in vivo senescence , 2015, Nature Reviews Cancer.

[9]  D. Beach,et al.  Ageing as developmental decay: insights from p16(INK4a.). , 2014, Trends in molecular medicine.

[10]  J. Deursen The role of senescent cells in ageing , 2014, Nature.

[11]  Byul A. Jee,et al.  Implications of time‐series gene expression profiles of replicative senescence , 2013, Aging cell.

[12]  S. Raguz,et al.  Co-regulation of senescence-associated genes by oncogenic homeobox proteins and polycomb repressive complexes , 2013, Cell cycle.

[13]  Juan M. Vaquerizas,et al.  DNA-Binding Specificities of Human Transcription Factors , 2013, Cell.

[14]  D. Baker,et al.  Senescent Cells: A Novel Therapeutic Target for Aging and Age‐Related Diseases , 2013, Clinical pharmacology and therapeutics.

[15]  S. Nevsimalova,et al.  Charcot–Marie–Tooth neuropathy due to a novel EGR2 gene mutation with mild phenotype – Usefulness of human mapping chip linkage analysis in a Czech family , 2012, Neuromuscular Disorders.

[16]  J. Campisi,et al.  Lamin B1 loss is a senescence-associated biomarker , 2012, Molecular biology of the cell.

[17]  Sunduz Keles,et al.  Genome-wide analysis of EGR2/SOX10 binding in myelinating peripheral nerve , 2012, Nucleic acids research.

[18]  N. LeBrasseur,et al.  Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders , 2011, Nature.

[19]  D. Beach,et al.  Primary cilium-dependent and -independent Hedgehog signaling inhibits p16(INK4A). , 2010, Molecular cell.

[20]  D. Beach,et al.  Multiple microRNAs rescue from Ras-induced senescence by inhibiting p21Waf1/Cip1 , 2010, Oncogene.

[21]  Anil Wipat,et al.  Feedback between p21 and reactive oxygen production is necessary for cell senescence , 2010, Molecular systems biology.

[22]  Heidi S Feiler,et al.  Molecular distinctions between stasis and telomere attrition senescence barriers shown by long-term culture of normal human mammary epithelial cells. , 2009, Cancer research.

[23]  J. Campisi,et al.  Persistent DNA damage signaling triggers senescence-associated inflammatory cytokine secretion , 2009, Nature Cell Biology.

[24]  J. Irelan,et al.  A Functional Screen for Regulators of CKDN2A Reveals MEOX2 as a Transcriptional Activator of INK4a , 2009, PloS one.

[25]  Judith Campisi,et al.  Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor , 2008, PLoS biology.

[26]  K. Ligon,et al.  p16INK4a induces an age-dependent decline in islet regenerative potential , 2006, Nature.

[27]  J. Vallat,et al.  Peripheral Myelin Maintenance Is a Dynamic Process Requiring Constant Krox20 Expression , 2006, The Journal of Neuroscience.

[28]  G. Peters,et al.  Regulation of the INK4b–ARF–INK4a tumour suppressor locus: all for one or one for all , 2006, Nature Reviews Molecular Cell Biology.

[29]  S. Werner,et al.  Cell cycle inhibitors p21 and p16 are required for the regulation of Schwann cell proliferation , 2006, Glia.

[30]  Kristjan R. Jessen,et al.  Krox-20 inhibits Jun-NH2-terminal kinase/c-Jun to control Schwann cell proliferation and death , 2004, The Journal of cell biology.

[31]  D. Beach,et al.  Polycomb CBX7 has a unifying role in cellular lifespan , 2004, Nature Cell Biology.

[32]  Yusuke Nakamura,et al.  Methylation at CpG islands in intron 1 of EGR2 confers enhancer‐like activity , 2003, FEBS letters.

[33]  Masashi Narita,et al.  Reversal of human cellular senescence: roles of the p53 and p16 pathways , 2003, The EMBO journal.

[34]  Dan Mercola,et al.  Early growth response 1 protein, an upstream gatekeeper of the p53 tumor suppressor, controls replicative senescence , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[35]  M. MacDonald,et al.  TBX-3, the Gene Mutated in Ulnar-Mammary Syndrome, Is a Negative Regulator of p19 ARF and Inhibits Senescence* , 2002, The Journal of Biological Chemistry.

[36]  G. Peters,et al.  Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence , 2001, Nature.

[37]  Thea D. Tlsty,et al.  Normal human mammary epithelial cells spontaneously escape senescence and acquire genomic changes , 2001, Nature.

[38]  Marc J. van de Vijver,et al.  Senescence bypass screen identifies TBX2, which represses Cdkn2a (p19ARF) and is amplified in a subset of human breast cancers , 2000, Nature Genetics.

[39]  Goberdhan P Dimri,et al.  Regulation of a Senescence Checkpoint Response by the E2F1 Transcription Factor and p14ARF Tumor Suppressor , 2000, Molecular and Cellular Biology.

[40]  N. Dyson The regulation of E2F by pRB-family proteins. , 1998, Genes & development.

[41]  Yue Xiong,et al.  ARF Promotes MDM2 Degradation and Stabilizes p53: ARF-INK4a Locus Deletion Impairs Both the Rb and p53 Tumor Suppression Pathways , 1998, Cell.

[42]  D. Wynford‐Thomas,et al.  Reinitiation of DNA Synthesis and Cell Division in Senescent Human Fibroblasts by Microinjection of Anti-p53 Antibodies , 1998, Molecular and Cellular Biology.

[43]  A. Beckmann,et al.  Egr transcription factors in the nervous system , 1997, Neurochemistry International.

[44]  S. Lowe,et al.  Oncogenic ras Provokes Premature Cell Senescence Associated with Accumulation of p53 and p16INK4a , 1997, Cell.

[45]  G. Hannon,et al.  Involvement of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of normal human fibroblasts. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[46]  S. Scherer,et al.  The Transcription Factors SCIP and Krox-20 Mark Distinct Stages and Cell Fates in Schwann Cell Differentiation , 1996, Molecular and Cellular Neuroscience.

[47]  S. Schneider-Maunoury,et al.  Krox-20 controls myelination in the peripheral nervous system , 1994, Nature.

[48]  L. Hayflick,et al.  The serial cultivation of human diploid cell strains. , 1961, Experimental cell research.