An integrative analysis reveals functional targets of GATA6 transcriptional regulation in gastric cancer

Lineage-restricted transcription factors (TFs) are frequently mutated or overexpressed in cancer and contribute toward malignant behaviors; however, the molecular bases of their oncogenic properties are largely unknown. As TF activities are difficult to inhibit directly with small molecules, the genes and pathways they regulate might represent more tractable targets for drug therapy. We studied GATA6, a TF gene that is frequently amplified or overexpressed in gastric, esophageal and pancreatic adenocarcinomas. GATA6-overexpressing gastric cancer cell lines cluster in gene expression space, separate from non-overexpressing lines. This expression clustering signifies a shared pathogenic group of genes that GATA6 may regulate through direct cis-element binding. We used chromatin immunoprecipitation and sequencing (ChIP-seq) to identify GATA6-bound genes and considered TF occupancy in relation to genes that respond to GATA6 depletion in cell lines and track with GATA6 mRNA (synexpression groups) in primary gastric cancers. Among other cellular functions, GATA6-occupied genes control apoptosis and govern the M-phase of the cell cycle. Depletion of GATA6 reduced the levels of the latter transcripts and arrested cells in G2 and M phases of the cell cycle. Synexpression in human tumor samples identified likely direct transcriptional targets substantially better than consideration only of transcripts that respond to GATA6 loss in cultured cells. Candidate target genes responded to the loss of GATA6 or its homolog GATA4 and even more to the depletion of both proteins. Many GATA6-dependent genes lacked nearby binding sites but several strongly dependent, synexpressed and GATA6-bound genes encode TFs such as MYC, HES1, RARB and CDX2. Thus, many downstream effects occur indirectly through other TFs and GATA6 activity in gastric cancer is partially redundant with GATA4. This integrative analysis of locus occupancy, gene dependency and synexpression provides a functional signature of GATA6-overexpressing gastric cancers, revealing both limits and new therapeutic directions for a challenging and frequently fatal disease.

[1]  X. Shirley Liu,et al.  Essential and Redundant Functions of Caudal Family Proteins in Activating Adult Intestinal Genes , 2011, Molecular and Cellular Biology.

[2]  Ryoichiro Kageyama,et al.  The Hes gene family: repressors and oscillators that orchestrate embryogenesis , 2007, Development.

[3]  A. Matsuda,et al.  Trends in stomach cancer mortality rates in Japan, USA, UK, France and Korea based on the WHO mortality database. , 2012, Japanese journal of clinical oncology.

[4]  T. Golub,et al.  Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma , 2005, Nature.

[5]  Hans Clevers,et al.  Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. , 2010, Cell stem cell.

[6]  Khay Guan Yeoh,et al.  A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets , 2012, Gut.

[7]  L. Olds,et al.  Spatio-temporal patterns of intestine-specific transcription factor expression during postnatal mouse gut development. , 2006, Gene expression patterns : GEP.

[8]  Benjamin J Bondow,et al.  Loss of both GATA4 and GATA6 blocks cardiac myocyte differentiation and results in acardia in mice. , 2008, Developmental biology.

[9]  D. Botstein,et al.  Variation in gene expression patterns in human gastric cancers. , 2003, Molecular biology of the cell.

[10]  M. Futai,et al.  Gastric DNA-binding proteins recognize upstream sequence motifs of parietal cell-specific genes. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[11]  Jorma Isola,et al.  In vivo amplification of the androgen receptor gene and progression of human prostate cancer , 1995, Nature Genetics.

[12]  J. Mesirov,et al.  Systematic investigation of genetic vulnerabilities across cancer cell lines reveals lineage-specific dependencies in ovarian cancer , 2011, Proceedings of the National Academy of Sciences.

[13]  D. Cunningham,et al.  Targeting the human EGFR family in esophagogastric cancer , 2011, Nature Reviews Clinical Oncology.

[14]  Reid F. Thompson,et al.  Widespread Hypomethylation Occurs Early and Synergizes with Gene Amplification during Esophageal Carcinogenesis , 2011, PLoS genetics.

[15]  R. Shivdasani,et al.  Notch signaling in stomach epithelial stem cell homeostasis , 2011, The Journal of experimental medicine.

[16]  C. Mathers,et al.  Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008 , 2010, International journal of cancer.

[17]  J. G. Park,et al.  Mutations in fibroblast growth factor receptor 2 and fibroblast growth factor receptor 3 genes associated with human gastric and colorectal cancers. , 2001, Cancer research.

[18]  R. Lucito,et al.  Frequent genomic copy number gain and overexpression of GATA-6 in pancreatic carcinoma , 2008, Cancer biology & therapy.

[19]  D. Frank Targeting transcription factors for cancer therapy. , 2008, IDrugs : the investigational drugs journal.

[20]  C. Mueller,et al.  GATA-4/5/6, a subfamily of three transcription factors transcribed in developing heart and gut. , 1994, The Journal of biological chemistry.

[21]  T. Hirao,et al.  The prognostic significance of amplification and overexpression of c‐met and c‐erb B‐2 in human gastric carcinomas , 1999, Cancer.

[22]  R. Playford,et al.  Reprogramming of intestinal differentiation and intercalary regeneration in Cdx2 mutant mice. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[23]  Ernest Fraenkel,et al.  Insights into GATA-1-mediated gene activation versus repression via genome-wide chromatin occupancy analysis. , 2009, Molecular cell.

[24]  Clifford A. Meyer,et al.  Differentiation-specific histone modifications reveal dynamic chromatin interactions and partners for the intestinal transcription factor CDX2. , 2010, Developmental cell.

[25]  M. Meyerson,et al.  Activation of GATA binding protein 6 (GATA6) sustains oncogenic lineage-survival in esophageal adenocarcinoma , 2012, Proceedings of the National Academy of Sciences.

[26]  M. van de Rijn,et al.  Genomic Profiling Identifies GATA6 as a Candidate Oncogene Amplified in Pancreatobiliary Cancer , 2008, PLoS genetics.

[27]  M. Meyerson,et al.  Gastrointestinal adenocarcinomas of the esophagus, stomach, and colon exhibit distinct patterns of genome instability and oncogenesis. , 2012, Cancer research.

[28]  Cem Elbi,et al.  FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. , 2008, Cancer research.

[29]  Yuriy L Orlov,et al.  The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. , 2010, Cell stem cell.

[30]  Elaine Dzierzak,et al.  Hes repressors are essential regulators of hematopoietic stem cell development downstream of Notch signaling , 2013, The Journal of experimental medicine.

[31]  E. Lander,et al.  Assessing the significance of chromosomal aberrations in cancer: Methodology and application to glioma , 2007, Proceedings of the National Academy of Sciences.

[32]  Manolis Kellis,et al.  Whole-genome ChIP-chip analysis of Dorsal, Twist, and Snail suggests integration of diverse patterning processes in the Drosophila embryo. , 2007, Genes & development.

[33]  Jeffrey W. Clark,et al.  MET amplification identifies a small and aggressive subgroup of esophagogastric adenocarcinoma with evidence of responsiveness to crizotinib. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[34]  Clifford A. Meyer,et al.  Cell-type selective chromatin remodeling defines the active subset of FOXA1-bound enhancers. , 2008, Genome research.

[35]  J. Darnell Transcription factors as targets for cancer therapy , 2002, Nature Reviews Cancer.

[36]  Heng Tao Shen,et al.  Principal Component Analysis , 2009, Encyclopedia of Biometrics.

[37]  A. Regev,et al.  SOX2 Is an Amplified Lineage Survival Oncogene in Lung and Esophageal Squamous Cell Carcinomas , 2009, Nature Genetics.

[38]  C. Allis,et al.  Extraction, purification and analysis of histones , 2007, Nature Protocols.

[39]  Hao Wang,et al.  Global regulation of erythroid gene expression by transcription factor GATA-1. , 2004, Blood.

[40]  Helga Thorvaldsdóttir,et al.  Integrative Genomics Viewer , 2011, Nature Biotechnology.

[41]  Yunbo Shi,et al.  Distinct Functions Are Implicated for the GATA-4, -5, and -6 Transcription Factors in the Regulation of Intestine Epithelial Cell Differentiation , 1998, Molecular and Cellular Biology.

[42]  E. Bresnick,et al.  GATA-1 Utilizes Ikaros and Polycomb Repressive Complex 2 To Suppress Hes1 and To Promote Erythropoiesis , 2012, Molecular and Cellular Biology.

[43]  Clifford A. Meyer,et al.  Cistrome: an integrative platform for transcriptional regulation studies , 2011, Genome Biology.

[44]  H. Clevers,et al.  Identification of stem cells in small intestine and colon by marker gene Lgr5 , 2007, Nature.

[45]  S. Duncan,et al.  GATA factors regulate proliferation, differentiation, and gene expression in small intestine of mature mice. , 2011, Gastroenterology.

[46]  T. Evans,et al.  Gata4 regulates the formation of multiple organs , 2005, Development.

[47]  Derek Y. Chiang,et al.  Characterizing the cancer genome in lung adenocarcinoma , 2007, Nature.

[48]  R. Poulsom,et al.  Identification of a metaplastic cell lineage associated with human gastric adenocarcinoma. , 1999, Laboratory investigation; a journal of technical methods and pathology.

[49]  Rafael A Irizarry,et al.  Exploration, normalization, and summaries of high density oligonucleotide array probe level data. , 2003, Biostatistics.