REST-DRD2 mechanism impacts glioblastoma stem cell-mediated tumorigenesis.

BACKGROUND Glioblastoma (GBM) is a lethal, heterogeneous human brain tumor, with regulatory mechanisms that have yet to be fully characterized. Previous studies have indicated that the transcriptional repressor REST (repressor element-1 silencing transcription factor) regulates the oncogenic potential of GBM stem cells (GSCs) based on level of expression. However, how REST performs its regulatory role is not well understood. METHODS We examined 2 independent high REST (HR) GSC lines using genome-wide assays, biochemical validations, gene knockdown analysis, and mouse tumor models. We analyzed in-house patient tumors and patient data present in The Cancer Genome Atlas (TCGA). RESULTS Genome-wide transcriptome and DNA-binding analyses suggested the dopamine receptor D2 (DRD2) gene, a dominant regulator of neurotransmitter signaling, as a direct target of REST. Biochemical analyses and mouse intracranial tumor models using knockdown of REST and double knockdown of REST and DRD2 validated this target and suggested that DRD2 is a downstream target of REST regulating tumorigenesis, at least in part, through controlling invasion and apoptosis. Further, TCGA GBM data support the presence of the REST-DRD2 axis and reveal that high REST/low DRD2 (HRLD) and low REST/high DRD2 (LRHD) tumors are specific subtypes, are molecularly different from the known GBM subtypes, and represent functional groups with distinctive patterns of enrichment of gene sets and biological pathways. The inverse HRLD/LRHD expression pattern is also seen in in-house GBM tumors. CONCLUSIONS These findings suggest that REST regulates neurotransmitter signaling pathways through DRD2 in HR-GSCs to impact tumorigenesis. They further suggest that the REST-DRD2 mechanism forms distinct subtypes of GBM.

[1]  G. Fuller,et al.  REST overexpression in mice causes deficits in spontaneous locomotion , 2018, Scientific Reports.

[2]  R. Zukin,et al.  REST, a master transcriptional regulator in neurodegenerative disease , 2018, Current Opinion in Neurobiology.

[3]  M. Weller,et al.  Vaccine-based immunotherapeutic approaches to gliomas and beyond , 2017, Nature Reviews Neurology.

[4]  C. Coarfa,et al.  REST represses miR-124 and miR-203 to regulate distinct oncogenic properties of glioblastoma stem cells , 2016, Neuro-oncology.

[5]  E. Chiocca,et al.  Glioma and microglia, a double entendre , 2016, Nature Immunology.

[6]  P. Tong,et al.  An expression based REST signature predicts patient survival and therapeutic response for glioblastoma multiforme , 2016, Scientific Reports.

[7]  Y. Nie,et al.  Dopamine D2 receptor suppresses gastric cancer cell invasion and migration via inhibition of EGFR/AKT/MMP-13 pathway. , 2016, International immunopharmacology.

[8]  G. Reifenberger,et al.  The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary , 2016, Acta Neuropathologica.

[9]  L. Marchionni,et al.  Alterations in cellular metabolome after pharmacological inhibition of Notch in glioblastoma cells , 2016, International journal of cancer.

[10]  Patrick Y. Wen,et al.  Neuro-oncology in 2015: Progress in glioma diagnosis, classification and treatment , 2016, Nature Reviews Neurology.

[11]  W. Wurst,et al.  The REST remodeling complex protects genomic integrity during embryonic neurogenesis , 2016, eLife.

[12]  K. Aldape,et al.  Mir-21–Sox2 Axis Delineates Glioblastoma Subtypes with Prognostic Impact , 2015, The Journal of Neuroscience.

[13]  J. Meldolesi,et al.  The Transcription Repressor REST in Adult Neurons: Physiology, Pathology, and Diseases,, , 2015, eNeuro.

[14]  J. Rich,et al.  Cancer stem cells in glioblastoma , 2015, Genes & development.

[15]  P. Pandolfi,et al.  TSPYL2 is an essential component of the REST/NRSF transcriptional complex for TGFβ signaling activation , 2015, Cell Death and Differentiation.

[16]  William A. Flavahan,et al.  Molecular targeting of TRF2 suppresses the growth and tumorigenesis of glioblastoma stem cells , 2014, Glia.

[17]  W. Cavenee,et al.  Genome-wide shRNA screen revealed integrated mitogenic signaling between dopamine receptor D2 (DRD2) and epidermal growth factor receptor (EGFR) in glioblastoma , 2014, Oncotarget.

[18]  D. Haussler,et al.  The Somatic Genomic Landscape of Glioblastoma , 2013, Cell.

[19]  E. Cattaneo,et al.  Binding of the repressor complex REST‐mSIN3b by small molecules restores neuronal gene transcription in Huntington's disease models , 2013, Journal of neurochemistry.

[20]  A. Roopra,et al.  A REST derived gene signature stratifies glioblastomas into chemotherapy resistant and responsive disease , 2012, BMC Genomics.

[21]  R. Dingledine,et al.  Epigenetics and epilepsy , 2012, Epilepsia.

[22]  Y. Ishikawa-Brush,et al.  Effects on promoter activity of common SNPs in 5′ region of GABRB3 exon 1A , 2012, Epilepsia.

[23]  T. MacDonald,et al.  REST Is a Novel Prognostic Factor and Therapeutic Target for Medulloblastoma , 2012, Molecular Cancer Therapeutics.

[24]  Tzong-Shiue Yu,et al.  A restricted cell population propagates glioblastoma growth following chemotherapy , 2012, Nature.

[25]  F. Zunino,et al.  REST Controls Self-Renewal and Tumorigenic Competence of Human Glioblastoma Cells , 2012, PloS one.

[26]  G. Fuller,et al.  REST regulates oncogenic properties of glioblastoma stem cells. , 2012, Stem cells.

[27]  M. Bennett,et al.  Repressor element-1 silencing transcription factor (REST)-dependent epigenetic remodeling is critical to ischemia-induced neuronal death , 2012, Proceedings of the National Academy of Sciences.

[28]  G. Fuller,et al.  Retinoic acid induces REST degradation and neuronal differentiation by modulating the expression of SCFβ‐TRCP in neuroblastoma cells , 2011, Cancer.

[29]  R. Gainetdinov,et al.  The Physiology, Signaling, and Pharmacology of Dopamine Receptors , 2011, Pharmacological Reviews.

[30]  K. Aldape,et al.  NFKBIA deletion in glioblastomas. , 2011, The New England journal of medicine.

[31]  J. Rees Neuro-oncology , 2010, Practical Neurology.

[32]  P. Dirks,et al.  Brain tumor stem cells: The cancer stem cell hypothesis writ large , 2010, Molecular oncology.

[33]  G. Fuller,et al.  Myoblast‐Derived Neuronal Cells Form Glutamatergic Neurons in the Mouse Cerebellum , 2010, Stem cells.

[34]  R. Wilson,et al.  Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. , 2010, Cancer cell.

[35]  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.

[36]  R. Prayson,et al.  The pathobiology of glioma tumors. , 2010, Annual review of pathology.

[37]  M. Rocchi,et al.  The Rest Repression of the Neurosecretory Phenotype Is Negatively Modulated by BHC80, a Protein of the BRAF/HDAC Complex , 2009, The Journal of Neuroscience.

[38]  J. Coulson,et al.  SCG3 Transcript in Peripheral Blood Is a Prognostic Biomarker for REST-Deficient Small Cell Lung Cancer , 2009, Clinical Cancer Research.

[39]  Thomas D. Wu,et al.  Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. , 2006, Cancer cell.

[40]  K. Aldape,et al.  Abnormal Expression of REST/NRSF and Myc in Neural Stem/Progenitor Cells Causes Cerebellar Tumors by Blocking Neuronal Differentiation , 2006, Molecular and Cellular Biology.

[41]  G. Mandel,et al.  The many faces of REST oversee epigenetic programming of neuronal genes , 2005, Current Opinion in Neurobiology.

[42]  R. Price,et al.  Many human medulloblastoma tumors overexpress repressor element-1 silencing transcription (REST)/neuron-restrictive silencer factor, which can be functionally countered by REST-VP16 , 2005, Molecular Cancer Therapeutics.

[43]  S. Kameoka,et al.  Activation of REST/NRSF Target Genes in Neural Stem Cells Is Sufficient To Cause Neuronal Differentiation , 2004, Molecular and Cellular Biology.

[44]  K. Aldape,et al.  Conversion of myoblasts to physiologically active neuronal phenotype. , 2004, Genes & development.

[45]  J. Morris,et al.  REST-VP16 activates multiple neuronal differentiation genes in human NT2 cells. , 2000, Nucleic acids research.

[46]  G. Fuller,et al.  The neuronal repressor REST/NRSF is an essential regulator in medulloblastoma cells , 2000, Nature Medicine.

[47]  Gail Mandel,et al.  REST: A mammalian silencer protein that restricts sodium channel gene expression to neurons , 1995, Cell.

[48]  D J Anderson,et al.  The neuron-restrictive silencer factor (NRSF): a coordinate repressor of multiple neuron-specific genes , 1995, Science.

[49]  R. Arceci Identification of a CpG Island Methylator Phenotype that Defines a Distinct Subgroup of Glioma , 2010 .

[50]  Mohamedi N. Kagalwala,et al.  Stemness is only a state of the cell. , 2008, Cold Spring Harbor symposia on quantitative biology.

[51]  A. Mortazavi,et al.  network : From single conserved sites to genome-wide repertoire Comparative genomics modeling of the NRSF / REST repressor Material Supplemental , 2006 .

[52]  J. Deuchars,et al.  Distinct profiles of REST interactions with its target genes at different stages of neuronal development. , 2005, Molecular biology of the cell.