Rare driver mutations in head and neck squamous cell carcinomas converge on NOTCH signaling

Cancer drivers converge on NOTCH Cancer genome–sequencing projects have emphasized the handful of genes mutated at high frequency in patients. Less attention has been directed to the hundreds of genes mutated in only a few patients—the so-called “long tail” mutations. Although rare, these mutations may nonetheless inform patient care. Loganathan et al. developed a reverse genetic CRISPR screen that allowed them to functionally assess in mice nearly 500 long tail gene mutations that occur in human head and neck squamous cell carcinoma (HNSCC). They identified 15 tumor-suppressor genes with activities that converged on the NOTCH signaling pathway. Given that NOTCH itself is mutated at high frequency in HNSCC, these results suggest that the growth of these tumors is largely driven by NOTCH inactivation. Science, this issue p. 1264 Functional analysis of rare mutations in head and neck cancers reveals that NOTCH signaling is disrupted in most tumors. In most human cancers, only a few genes are mutated at high frequencies; most are mutated at low frequencies. The functional consequences of these recurrent but infrequent “long tail” mutations are often unknown. We focused on 484 long tail genes in head and neck squamous cell carcinoma (HNSCC) and used in vivo CRISPR to screen for genes that, upon mutation, trigger tumor development in mice. Of the 15 tumor-suppressor genes identified, ADAM10 and AJUBA suppressed HNSCC in a haploinsufficient manner by promoting NOTCH receptor signaling. ADAM10 and AJUBA mutations or monoallelic loss occur in 28% of human HNSCC cases and are mutually exclusive with NOTCH receptor mutations. Our results show that oncogenic mutations in 67% of human HNSCC cases converge onto the NOTCH signaling pathway, making NOTCH inactivation a hallmark of HNSCC.

[1]  S. Henikoff,et al.  An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites , 2016, bioRxiv.

[2]  H. Ohtsuki,et al.  Accumulation of driver and passenger mutations during tumor progression , 2009, Proceedings of the National Academy of Sciences.

[3]  M. Bailly,et al.  Ajuba is required for Rac activation and maintenance of E-cadherin adhesion , 2011, The Journal of cell biology.

[4]  Amber L. Couzens,et al.  The CRAPome: a Contaminant Repository for Affinity Purification Mass Spectrometry Data , 2013, Nature Methods.

[5]  A. Gingras,et al.  Parallel Exploration of Interaction Space by BioID and Affinity Purification Coupled to Mass Spectrometry. , 2017, Methods in molecular biology.

[6]  Steven J. M. Jones,et al.  Comprehensive genomic characterization of head and neck squamous cell carcinomas , 2015, Nature.

[7]  T. Akiyama,et al.  Ajuba negatively regulates the Wnt signaling pathway by promoting GSK-3β-mediated phosphorylation of β-catenin , 2008, Oncogene.

[8]  M. Zenke,et al.  Activated Notch1 Target Genes during Embryonic Cell Differentiation Depend on the Cellular Context and Include Lineage Determinants and Inhibitors , 2010, PloS one.

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

[10]  A. Kulkarni,et al.  Loss of TGF-β signaling and PTEN promotes head and neck squamous cell carcinoma through cellular senescence evasion and cancer-related inflammation , 2012, Oncogene.

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

[12]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

[13]  K. Billiar,et al.  TRIP6 inhibits Hippo signaling in response to tension at adherens junctions , 2018, EMBO reports.

[14]  K. Guan,et al.  Angiomotin is a novel Hippo pathway component that inhibits YAP oncoprotein. , 2011, Genes & development.

[15]  E. Shoubridge,et al.  A proximity biotinylation map of a human cell , 2019, bioRxiv.

[16]  A. Rustgi,et al.  Squamous Cell Cancers: A Unified Perspective on Biology and Genetics. , 2016, Cancer cell.

[17]  N. Socci,et al.  Synthetic lethality in ATM-deficient RAD50-mutant tumors underlies outlier response to cancer therapy. , 2014, Cancer discovery.

[18]  G. Longmore,et al.  Ajuba LIM Proteins Are Negative Regulators of the Hippo Signaling Pathway , 2010, Current Biology.

[19]  I. Tomlinson,et al.  The mini-driver model of polygenic cancer evolution , 2015, Nature Reviews Cancer.

[20]  Rohit Reja,et al.  The RIPK4–IRF6 signalling axis safeguards epidermal differentiation and barrier function , 2019, Nature.

[21]  Anshul Kundaje,et al.  The ENCODE Blacklist: Identification of Problematic Regions of the Genome , 2019, Scientific Reports.

[22]  M. Duffy,et al.  The ADAMs family of proteases as targets for the treatment of cancer , 2016, Cancer biology & therapy.

[23]  Neville E. Sanjana,et al.  Improved vectors and genome-wide libraries for CRISPR screening , 2014, Nature Methods.

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

[25]  S. Egan,et al.  Cooperation between Pik3ca and p53 mutations in mouse mammary tumor formation. , 2011, Cancer research.

[26]  Joshua M. Stuart,et al.  Resource Genomic , Pathway Networ k , and Immunologic Features Distinguishing Squamous Carcinomas Graphical , 2018 .

[27]  C. Blobel,et al.  The disintegrin/metalloproteinase Adam10 is essential for epidermal integrity and Notch-mediated signaling , 2011, Development.

[28]  Yi Zhou,et al.  A CRISPR/Cas9 vector system for tissue-specific gene disruption in zebrafish. , 2015, Developmental cell.

[29]  E. Shoubridge,et al.  A proximity-dependent biotinylation map of a human cell , 2021, Nature.

[30]  C. McGlade,et al.  Mammalian Numb Proteins Promote Notch1 Receptor Ubiquitination and Degradation of the Notch1 Intracellular Domain* , 2003, Journal of Biological Chemistry.

[31]  H. Saya,et al.  Aurora-A and an Interacting Activator, the LIM Protein Ajuba, Are Required for Mitotic Commitment in Human Cells , 2003, Cell.

[32]  Tony Pawson,et al.  Protein Interaction Network of the Mammalian Hippo Pathway Reveals Mechanisms of Kinase-Phosphatase Interactions , 2013, Science Signaling.

[33]  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.

[34]  Ruud H. Brakenhoff,et al.  The molecular landscape of head and neck cancer , 2018, Nature Reviews Cancer.

[35]  G. Weskamp,et al.  Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of six EGFR ligands , 2004, The Journal of cell biology.

[36]  S. Molyneux,et al.  Notch activation by the metalloproteinase ADAM17 regulates myeloproliferation and atopic barrier immunity by suppressing epithelial cytokine synthesis. , 2012, Immunity.

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

[38]  E. Wagner,et al.  Differentiation-induced skin cancer suppression by FOS, p53, and TACE/ADAM17. , 2012, The Journal of clinical investigation.

[39]  D. Durocher,et al.  High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities , 2015, Cell.

[40]  Steven J. M. Jones,et al.  Oncogenic Signaling Pathways in The Cancer Genome Atlas. , 2018, Cell.

[41]  Elaine Fuchs,et al.  Direct in Vivo RNAi Screen Unveils Myosin IIa as a Tumor Suppressor of Squamous Cell Carcinomas , 2014, Science.

[42]  E. Lander,et al.  Lessons from the Cancer Genome , 2013, Cell.

[43]  E. Fuchs,et al.  Rapid functional dissection of genetic networks via tissue-specific transduction and RNAi in mouse embryos , 2010, Nature Medicine.

[44]  K. Kinzler,et al.  Cancer Genome Landscapes , 2013, Science.

[45]  F. Radtke,et al.  Notch as a tumour suppressor , 2017, Nature Reviews Cancer.

[46]  E. Fuchs,et al.  RNAi-mediated gene function analysis in skin. , 2013, Methods in molecular biology.

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

[48]  A. Emili,et al.  DLG5 connects cell polarity and Hippo signaling protein networks by linking PAR-1 with MST1/2 , 2016, Genes & development.

[49]  C. Sander,et al.  Genome Sequencing Identifies a Basis for Everolimus Sensitivity , 2012, Science.

[50]  Junjie Chen,et al.  Angiomotin-like Proteins Associate with and Negatively Regulate YAP1* , 2010, The Journal of Biological Chemistry.