Transposon mutagenesis identifies genes that transform neural stem cells into glioma-initiating cells

Neural stem cells (NSCs) are considered to be the cell of origin of glioblastoma multiforme (GBM). However, the genetic alterations that transform NSCs into glioma-initiating cells remain elusive. Using a unique transposon mutagenesis strategy that mutagenizes NSCs in culture, followed by additional rounds of mutagenesis to generate tumors in vivo, we have identified genes and signaling pathways that can transform NSCs into glioma-initiating cells. Mobilization of Sleeping Beauty transposons in NSCs induced the immortalization of astroglial-like cells, which were then able to generate tumors with characteristics of the mesenchymal subtype of GBM on transplantation, consistent with a potential astroglial origin for mesenchymal GBM. Sequence analysis of transposon insertion sites from tumors and immortalized cells identified more than 200 frequently mutated genes, including human GBM-associated genes, such as Met and Nf1, and made it possible to discriminate between genes that function during astroglial immortalization vs. later stages of tumor development. We also functionally validated five GBM candidate genes using a previously undescribed high-throughput method. Finally, we show that even clonally related tumors derived from the same immortalized line have acquired distinct combinations of genetic alterations during tumor development, suggesting that tumor formation in this model system involves competition among genetically variant cells, which is similar to the Darwinian evolutionary processes now thought to generate many human cancers. This mutagenesis strategy is faster and simpler than conventional transposon screens and can potentially be applied to any tissue stem/progenitor cells that can be grown and differentiated in vitro.

[1]  C. Doe Neural stem cells: balancing self-renewal with differentiation , 2008, Development.

[2]  Joshua M. Korn,et al.  Comprehensive genomic characterization defines human glioblastoma genes and core pathways , 2008, Nature.

[3]  D. Ingram,et al.  Neurofibroma-associated growth factors activate a distinct signaling network to alter the function of neurofibromin-deficient endothelial cells. , 2006, Human molecular genetics.

[4]  D. Steindler,et al.  Identification of a multipotent astrocytic stem cell in the immature and adult mouse brain. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[5]  E. Shaulian,et al.  Identification of a minimal transforming domain of p53: negative dominance through abrogation of sequence-specific DNA binding , 1992, Molecular and cellular biology.

[6]  T. Jacks,et al.  Mutant p53 Gain of Function in Two Mouse Models of Li-Fraumeni Syndrome , 2004, Cell.

[7]  C. Maley,et al.  Cancer is a disease of clonal evolution within the body1–3. This has profound clinical implications for neoplastic progression, cancer prevention and cancer therapy. Although the idea of cancer as an evolutionary problem , 2006 .

[8]  R. DePinho,et al.  The differential impact of p16INK4a or p19ARF deficiency on cell growth and tumorigenesis , 2004, Oncogene.

[9]  R. DePinho,et al.  Epidermal growth factor receptor and Ink4a/Arf: convergent mechanisms governing terminal differentiation and transformation along the neural stem cell to astrocyte axis. , 2002, Cancer cell.

[10]  N. Copeland,et al.  Insertional mutagenesis identifies genes that promote the immortalization of primary bone marrow progenitor cells. , 2005, Blood.

[11]  T. Scheetz,et al.  A modified sleeping beauty transposon system that can be used to model a wide variety of human cancers in mice. , 2009, Cancer research.

[12]  P. A. Futreal,et al.  Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. , 2012, The New England journal of medicine.

[13]  Chunxu Qu,et al.  Cooperativity within and among Pten, p53, and Rb pathways induces high-grade astrocytoma in adult brain. , 2011, Cancer cell.

[14]  K. Anderson,et al.  Genetic variegation of clonal architecture and propagating cells in leukaemia , 2011, Nature.

[15]  Lincoln D. Stein,et al.  Identification of a therapeutic strategy targeting amplified FGF19 in liver cancer by Oncogenomic screening. , 2011, Cancer cell.

[16]  Jason A. Koutcher,et al.  Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis , 2005, Nature.

[17]  O. Kretz,et al.  Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety , 1999, Nature Genetics.

[18]  S. Minucci,et al.  Overexpression of sPRDM16 coupled with loss of p53 induces myeloid leukemias in mice. , 2007, The Journal of clinical investigation.

[19]  A. Rust,et al.  Sleeping Beauty mutagenesis reveals cooperating mutations and pathways in pancreatic adenocarcinoma , 2012, Proceedings of the National Academy of Sciences.

[20]  I. Weissman,et al.  Stem cells, cancer, and cancer stem cells , 2001, Nature.

[21]  Corey M. Carlson,et al.  Cancer gene discovery in solid tumours using transposon-based somatic mutagenesis in the mouse , 2005, Nature.

[22]  P. Carlsson,et al.  Foxj3, a novel mammalian forkhead gene expressed in neuroectoderm, neural crest, and myotome , 2004, Developmental dynamics : an official publication of the American Association of Anatomists.

[23]  S. Gabriel,et al.  Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. , 2010, Cancer cell.

[24]  R. Henkelman,et al.  Identification of human brain tumour initiating cells , 2004, Nature.

[25]  A. Multani,et al.  Chromosome stability, in the absence of apoptosis, is critical for suppression of tumorigenesis in Trp53 mutant mice , 2004, Nature Genetics.

[26]  Jie Peng,et al.  Isolation and culture of rat and mouse oligodendrocyte precursor cells , 2007, Nature Protocols.

[27]  S. Weiss,et al.  Clonal and population analyses demonstrate that an EGF-responsive mammalian embryonic CNS precursor is a stem cell. , 1996, Developmental biology.

[28]  Jaap Kool,et al.  High throughput insertional mutagenesis screens in mice to identify oncogenic networks , 2009, Nature Reviews Cancer.

[29]  A. Rust,et al.  Insertional mutagenesis identifies multiple networks of co-operating genes driving intestinal tumorigenesis , 2011, Nature Genetics.

[30]  G. Bishop,et al.  Tumor necrosis factor receptor-associated factor 3 is a critical regulator of B cell homeostasis in secondary lymphoid organs. , 2007, Immunity.

[31]  Laurence Ettwiller,et al.  Large-scale analysis of the regulatory architecture of the mouse genome with a transposon-associated sensor , 2011, Nature Genetics.

[32]  S. Carmichael,et al.  Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neurogenesis , 2009, The Journal of Neuroscience.

[33]  Y. Xing,et al.  A Transcriptome Database for Astrocytes, Neurons, and Oligodendrocytes: A New Resource for Understanding Brain Development and Function , 2008, The Journal of Neuroscience.

[34]  E. Cuppen,et al.  High-throughput semiquantitative analysis of insertional mutations in heterogeneous tumors. , 2011, Genome research.

[35]  N. Copeland,et al.  Harnessing transposons for cancer gene discovery , 2010, Nature Reviews Cancer.

[36]  P. Kleihues,et al.  Epidemiology and etiology of gliomas , 2005, Acta Neuropathologica.

[37]  L. Parada,et al.  Neurofibromin, a tumor suppressor in the nervous system. , 2001, Experimental cell research.

[38]  D. Rowitch,et al.  Glioma Stem Cells: A Midterm Exam , 2008, Neuron.

[39]  S. Scherer,et al.  Clonal Selection Drives Genetic Divergence of Metastatic Medulloblastoma , 2012, Nature.

[40]  M. Stratton,et al.  COSMIC 2005 , 2006, British Journal of Cancer.

[41]  Hongtao Yu,et al.  Mutational Inactivation of STAG2 Causes Aneuploidy in Human Cancer , 2011, Science.

[42]  H. Armah Malignant Astrocytomas Originate from Neural Stem/Progenitor Cells in a Somatic Tumor Suppressor Mouse Model , 2010 .

[43]  A. Krasnitz,et al.  An Oncogenomics-Based In Vivo RNAi Screen Identifies Tumor Suppressors in Liver Cancer , 2008, Cell.

[44]  S. Ogawa,et al.  Evi-1 is a critical regulator for hematopoietic stem cells and transformed leukemic cells. , 2008, Cell stem cell.

[45]  A. V. van Rossum,et al.  Ablation of the retinoblastoma gene family deregulates G(1) control causing immortalization and increased cell turnover under growth-restricting conditions. , 2000, Genes & development.

[46]  D. Largaespada,et al.  Mammalian mutagenesis using a highly mobile somatic Sleeping Beauty transposon system , 2005, Nature.

[47]  Michael A Choti,et al.  Inactivating mutations of the chromatin remodeling gene ARID2 in hepatocellular carcinoma , 2011, Nature Genetics.

[48]  L. Luo,et al.  Mosaic Analysis with Double Markers Reveals Tumor Cell of Origin in Glioma , 2011, Cell.

[49]  Miriam Scadeng,et al.  Development of a novel mouse glioma model using lentiviral vectors , 2009, Nature Medicine.

[50]  J. Testa,et al.  FAS-Associated Factor 1 (FAF1): Diverse functions and implications for oncogenesis , 2009, Cell cycle.

[51]  H. Varmus,et al.  Modeling mutations in the G1 arrest pathway in human gliomas: overexpression of CDK4 but not loss of INK4a-ARF induces hyperploidy in cultured mouse astrocytes. , 1998, Genes & development.

[52]  D. Busam,et al.  An Integrated Genomic Analysis of Human Glioblastoma Multiforme , 2008, Science.

[53]  J. Milner,et al.  Cotranslation of activated mutant p53 with wild type drives the wild-type p53 protein into the mutant conformation , 1991, Cell.

[54]  Jane E. Visvader,et al.  Cells of origin in cancer , 2011, Nature.

[55]  Philip A Beachy,et al.  Hedgehog-Regulated Processing of Gli3 Produces an Anterior/Posterior Repressor Gradient in the Developing Vertebrate Limb , 2000, Cell.

[56]  A. Álvarez-Buylla,et al.  Neural stem cells in mammalian development. , 2006, Current opinion in cell biology.