IER5, a DNA-damage response gene, is required for Notch-mediated induction of squamous cell differentiation

Notch signaling regulates normal squamous cell proliferation and differentiation and is frequently disrupted in squamous cell carcinomas, in which Notch is a key tumor suppressive pathway. To identify the direct targets of Notch that produce these phenotypes, we introduced a conditional Notch transgene into squamous cell carcinoma cell lines, which respond to Notch activation in 2D culture and in organoid cultures by undergoing differentiation. RNA-seq and ChIP-seq analyses show that in squamous cells Notch activates a context-specific program of gene expression that depends on lineage-specific regulatory elements, most of which lie in long- range enhancers. Among the direct Notch target genes are multiple DNA damage response genes, including IER5, which is regulated by Notch through several enhancer elements. We show that IER5 is required for Notch-induced differentiation in squamous carcinoma cells and in TERT-immortalized keratinocytes. Its function is epistatic to PPP2R2A, which encodes the B55α subunit of PP2A, and IER5 interacts with B55α in cells and in purified systems. These results show that Notch and DNA-damage response pathways converge in squamous cells and that some components of these pathways promote differentiation, which may serve to eliminate DNA-damaged cells from the proliferative pool in squamous epithelia. Crosstalk involving Notch and PP2A may enable Notch signaling to be tuned and integrated with other pathways that regulate squamous differentiation. Our work also suggests that squamous cell carcinomas remain responsive to Notch signaling, providing a rationale for reactivation of Notch as a therapeutic strategy in these cancers. Impact Our findings highlight context-specific crosstalk between Notch, DNA damage response genes, and PP2A, and provide a roadmap for understanding how Notch induces the differentiation of squamous cells.

[1]  Anne-Claude Gingras,et al.  Rare driver mutations in head and neck squamous cell carcinomas converge on NOTCH signaling , 2020, Science.

[2]  D. Sprinzak,et al.  Exposure to Mites Sensitizes Intestinal Stem Cell Maintenance, Splenic Marginal Zone B Cell Homeostasis, And Heart Development to Notch Dosage and Cooperativity , 2020, bioRxiv.

[3]  J. Aster,et al.  Extension of the Notch intracellular domain ankyrin repeat stack by NRARP promotes feedback inhibition of Notch signaling , 2019, Science Signaling.

[4]  P. Jones,et al.  Outcompeting p53-Mutant Cells in the Normal Esophagus by Redox Manipulation , 2019, Cell stem cell.

[5]  Brijesh K. Garg,et al.  Tandem Affinity Purification and Mass Spectrometry (TAP‐MS) for the Analysis of Protein Complexes , 2019, Current protocols in protein science.

[6]  Shawn C. Little,et al.  Oncogenic Notch promotes long-range regulatory interactions within hyperconnected 3D cliques , 2019, bioRxiv.

[7]  Hernan G. Garcia,et al.  Enhancer priming enables fast and sustained transcriptional responses to Notch signaling , 2018, bioRxiv.

[8]  M. Stratton,et al.  The genomic landscape of cutaneous SCC reveals drivers and a novel azathioprine associated mutational signature , 2018, Nature Communications.

[9]  Gordon K Smyth,et al.  The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads , 2018, bioRxiv.

[10]  Shawn M. Gillespie,et al.  A B Cell Regulome Links Notch to Downstream Oncogenic Pathways in Small B Cell Lymphomas. , 2017, Cell reports.

[11]  L. Di Marcotullio,et al.  Maml1 acts cooperatively with Gli proteins to regulate sonic hedgehog signaling pathway , 2017, Cell Death & Disease.

[12]  J. Aster,et al.  Genome-wide identification and characterization of Notch transcription complex–binding sequence-paired sites in leukemia cells , 2017, Science Signaling.

[13]  J. Aster,et al.  The Varied Roles of Notch in Cancer. , 2017, Annual review of pathology.

[14]  M. Grace,et al.  Cutaneous HPV8 and MmuPV1 E6 Proteins Target the NOTCH and TGF-β Tumor Suppressors to Inhibit Differentiation and Sustain Keratinocyte Proliferation , 2017, PLoS pathogens.

[15]  Kara Dolinski,et al.  The BioGRID interaction database: 2017 update , 2016, Nucleic Acids Res..

[16]  S. Bray Notch signalling in context , 2016, Nature Reviews Molecular Cell Biology.

[17]  H. Jiang,et al.  Transcriptional regulation of IER5 in response to radiation in HepG2 , 2016, Cancer Gene Therapy.

[18]  H. Aburatani,et al.  IER5 generates a novel hypo-phosphorylated active form of HSF1 and contributes to tumorigenesis , 2016, Scientific Reports.

[19]  Shawn M. Gillespie,et al.  An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma , 2016, Nature Genetics.

[20]  K. Green,et al.  In Vitro Model of the Epidermis: Connecting Protein Function to 3D Structure. , 2016, Methods in enzymology.

[21]  H. Sakurai,et al.  Immediate‐early response 5 (IER5) interacts with protein phosphatase 2A and regulates the phosphorylation of ribosomal protein S6 kinase and heat shock factor 1 , 2015, FEBS letters.

[22]  S. Russell,et al.  Chromatin signatures at Notch‐regulated enhancers reveal large‐scale changes in H3K56ac upon activation , 2015, The EMBO journal.

[23]  H. Sakurai,et al.  HSF1 transcriptional activity is modulated by IER5 and PP2A/B55 , 2015, FEBS letters.

[24]  H. Sakurai,et al.  Heat‐induced expression of the immediate‐early gene IER5 and its involvement in the proliferation of heat‐shocked cells , 2015, The FEBS journal.

[25]  Zhenyi Liu,et al.  The intracellular domains of Notch1 and Notch2 are functionally equivalent during development and carcinogenesis , 2015, Development.

[26]  W. Hahn,et al.  BreaKmer: detection of structural variation in targeted massively parallel sequencing data using kmers , 2014, Nucleic acids research.

[27]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[28]  M. Sweet,et al.  Receptor-interacting Protein Kinase 4 and Interferon Regulatory Factor 6 Function as a Signaling Axis to Regulate Keratinocyte Differentiation* , 2014, The Journal of Biological Chemistry.

[29]  S. Dudoit,et al.  Normalization of RNA-seq data using factor analysis of control genes or samples , 2014, Nature Biotechnology.

[30]  Philip Greulich,et al.  Differentiation imbalance in single oesophageal progenitor cells causes clonal immortalization and field change , 2014, Nature Cell Biology.

[31]  Eric Nestler,et al.  ngs.plot: Quick mining and visualization of next-generation sequencing data by integrating genomic databases , 2014, BMC Genomics.

[32]  S. Arron,et al.  NOTCH1 mutations occur early during cutaneous squamous cell carcinogenesis , 2014, The Journal of investigative dermatology.

[33]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[34]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[35]  Jon C. Aster,et al.  NOTCH1–RBPJ complexes drive target gene expression through dynamic interactions with superenhancers , 2013, Proceedings of the National Academy of Sciences.

[36]  Allon M. Klein,et al.  Interfollicular Epidermal Stem Cells Self-Renew via Autocrine Wnt Signaling , 2013, Science.

[37]  H. Stunnenberg,et al.  Dynamic binding of RBPJ is determined by Notch signaling status. , 2013, Genes & development.

[38]  A. Sivachenko,et al.  Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples , 2013, Nature Biotechnology.

[39]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[40]  Yuchen Jiao,et al.  Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. , 2012, Cancer discovery.

[41]  James T. Webber,et al.  Interpreting cancer genomes using systematic host network perturbations by tumour virus proteins - eScholarship , 2012 .

[42]  James T. Webber,et al.  Interpreting cancer genomes using systematic host perturbations by tumour virus proteins , 2012, Nature.

[43]  J. Aster,et al.  Targeting the Notch pathway: twists and turns on the road to rational therapeutics. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[44]  J. Aster,et al.  Cutaneous β-human papillomavirus E6 proteins bind Mastermind-like coactivators and repress Notch signaling , 2012, Proceedings of the National Academy of Sciences.

[45]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[46]  Nikhil Wagle,et al.  High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. , 2012, Cancer discovery.

[47]  G. Dotto,et al.  IRF6 is a mediator of Notch pro‐differentiation and tumour suppressive function in keratinocytes , 2011, The EMBO journal.

[48]  David Haussler,et al.  Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma , 2011, Proceedings of the National Academy of Sciences.

[49]  R. Gibbs,et al.  Exome Sequencing of Head and Neck Squamous Cell Carcinoma Reveals Inactivating Mutations in NOTCH1 , 2011, Science.

[50]  Philip Machanick,et al.  MEME-ChIP: motif analysis of large DNA datasets , 2011, Bioinform..

[51]  M. DePristo,et al.  A framework for variation discovery and genotyping using next-generation DNA sequencing data , 2011, Nature Genetics.

[52]  K. Purdie,et al.  Isolation and culture of squamous cell carcinoma lines. , 2011, Methods in molecular biology.

[53]  J. Aster,et al.  Structural and mechanistic insights into cooperative assembly of dimeric Notch transcription complexes , 2010, Nature Structural &Molecular Biology.

[54]  M. DePristo,et al.  The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. , 2010, Genome research.

[55]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[56]  J. Griffin,et al.  The Mastermind-like 1 (MAML1) Co-activator Regulates Constitutive NF-κB Signaling and Cell Survival* , 2010, The Journal of Biological Chemistry.

[57]  Aaron R. Quinlan,et al.  BEDTools: a flexible suite of utilities for comparing genomic features , 2010, Bioinform..

[58]  April Bezdek Pomey RhoV, a Potential Notch Target Gene in Murine Skin, Does not Play an Essential Role in Epidermis Development and Homeostasis , 2010 .

[59]  Yi Zhang,et al.  multiplierz: an extensible API based desktop environment for proteomics data analysis , 2009, BMC Bioinformatics.

[60]  D. Hunter,et al.  mixtools: An R Package for Analyzing Mixture Models , 2009 .

[61]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[62]  Jignesh R. Parikh,et al.  mzAPI: a new strategy for efficiently sharing mass spectrometry data , 2009, Nature Methods.

[63]  Yu Wang,et al.  Induced expression of the IER5 gene by γ-ray irradiation and its involvement in cell cycle checkpoint control and survival , 2009, Radiation and environmental biophysics.

[64]  J. Aster,et al.  Structure of the Notch1-negative regulatory region: implications for normal activation and pathogenic signaling in T-ALL. , 2008, Blood.

[65]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[66]  Clifford A. Meyer,et al.  Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.

[67]  J. Aster,et al.  Leukemia-associated NOTCH1 alleles are weak tumor initiators but accelerate K-ras-initiated leukemia. , 2008, The Journal of clinical investigation.

[68]  G. Dotto,et al.  The FoxO3a gene is a key negative target of canonical Notch signalling in the keratinocyte UVB response , 2008, The EMBO journal.

[69]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[70]  S. Bray,et al.  Notch activation stimulates transient and selective binding of Su(H)/CSL to target enhancers. , 2007, Genes & development.

[71]  Goberdhan P Dimri,et al.  The Notch Regulator MAML1 Interacts with p53 and Functions as a Coactivator* , 2007, Journal of Biological Chemistry.

[72]  Piotr Sliz,et al.  Cooperative assembly of higher-order Notch complexes functions as a switch to induce transcription , 2007, Proceedings of the National Academy of Sciences.

[73]  A. Falus,et al.  Microarray analysis of radiation response genes in primary human fibroblasts. , 2006, International journal of radiation oncology, biology, physics.

[74]  Elaine Fuchs,et al.  Canonical notch signaling functions as a commitment switch in the epidermal lineage. , 2006, Genes & development.

[75]  Fiona M Watt,et al.  Single-cell expression profiling of human epidermal stem and transit-amplifying cells: Lrig1 is a regulator of stem cell quiescence , 2006, Proceedings of the National Academy of Sciences.

[76]  J. Aster,et al.  c-Myc is an important direct target of Notch1 in T-cell acute lymphoblastic leukemia/lymphoma. , 2006, Genes & development.

[77]  W. Pear,et al.  Impaired notch signaling promotes de novo squamous cell carcinoma formation. , 2006, Cancer research.

[78]  C. Simbulan-Rosenthal,et al.  Id3 induces a caspase-3- and -9-dependent apoptosis and mediates UVB sensitization of HPV16 E6/7 immortalized human keratinocytes , 2006, Oncogene.

[79]  J. Aster,et al.  Leukemia-Associated Mutations within the NOTCH1 Heterodimerization Domain Fall into at Least Two Distinct Mechanistic Classes , 2006, Molecular and Cellular Biology.

[80]  R. Bronson,et al.  The Notch coactivator, MAML1, functions as a novel coactivator for MEF2C-mediated transcription and is required for normal myogenesis. , 2006, Genes & development.

[81]  Jon C. Aster,et al.  Structural Basis for Cooperativity in Recruitment of MAML Coactivators to Notch Transcription Complexes , 2006, Cell.

[82]  L. Hooper Faculty Opinions recommendation of Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. , 2005 .

[83]  A. Christiano,et al.  Activation of Notch1 in the hair follicle leads to cell-fate switch and Mohawk alopecia. , 2004, Differentiation; research in biological diversity.

[84]  G. Gibson,et al.  Microarray Analysis , 2020, Definitions.

[85]  Hans Clevers,et al.  Notch1 functions as a tumor suppressor in mouse skin , 2003, Nature Genetics.

[86]  James D. Griffin,et al.  Growth Suppression of Pre-T Acute Lymphoblastic Leukemia Cells by Inhibition of Notch Signaling , 2003, Molecular and Cellular Biology.

[87]  K. Münger,et al.  Abrogation of the retinoblastoma tumor suppressor checkpoint during keratinocyte immortalization is not sufficient for induction of centrosome-mediated genomic instability. , 2003, Cancer research.

[88]  N. Perrimon,et al.  γ‐Secretase/presenilin inhibitors for Alzheimer's disease phenocopy Notch mutations in Drosophila , 2003, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[89]  H. Hirata,et al.  Oscillatory Expression of the bHLH Factor Hes1 Regulated by a Negative Feedback Loop , 2002, Science.

[90]  J-Z Qin,et al.  Jagged-1 mediated activation of notch signaling induces complete maturation of human keratinocytes through NF-κB and PPARγ , 2002, Cell Death and Differentiation.

[91]  V. Tron,et al.  GADD45 regulates G2/M arrest, DNA repair, and cell death in keratinocytes following ultraviolet exposure. , 2002, The Journal of investigative dermatology.

[92]  J. Jorcano,et al.  Analysis of the ultraviolet B response in primary human keratinocytes using oligonucleotide microarrays , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[93]  J-Z Qin,et al.  Jagged-1 mediated activation of notch signaling induces complete maturation of human keratinocytes through NF-kappaB and PPARgamma. , 2002, Cell death and differentiation.

[94]  Freddy Radtke,et al.  Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation , 2001, The EMBO journal.

[95]  I. Bernstein,et al.  Immobilization of Notch ligand, Delta-1, is required for induction of notch signaling. , 2000, Journal of cell science.

[96]  F. Watt,et al.  Stimulation of human epidermal differentiation by Delta–Notch signalling at the boundaries of stem-cell clusters , 2000, Current Biology.

[97]  J. Sklar,et al.  Calcium Depletion Dissociates and Activates Heterodimeric Notch Receptors , 2000, Molecular and Cellular Biology.

[98]  B. Birren,et al.  Ier5, a novel member of the slow-kinetics immediate-early genes. , 1999, Genomics.

[99]  R. Kumar,et al.  A novel immediate early response gene, IEX-1, is induced by ultraviolet radiation in human keratinocytes. , 1998, Biochemical and biophysical research communications.

[100]  S. Artavanis-Tsakonas,et al.  Secreted forms of DELTA and SERRATE define antagonists of Notch signaling in Drosophila. , 1997, Development.

[101]  F. Di Cunto,et al.  The absence of p21Cip1/WAF1 alters keratinocyte growth and differentiation and promotes ras-tumor progression. , 1996, Genes & development.

[102]  R. Berger,et al.  Identification of BTG2, an antiproliferative p53–dependent component of the DNA damage cellular response pathway , 1996, Nature Genetics.

[103]  E. Fuchs Keratins and the skin. , 1995, Annual review of cell and developmental biology.

[104]  Fiona M. Watt,et al.  Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression , 1993, Cell.

[105]  E. McCall Calcium Depletion , 1936 .