TET2 is required to suppress mTORC1 signaling through urea cycle with therapeutic potential

[1]  Ruonan Zhang,et al.  Targeting fibrinogen-like protein 1 enhances immunotherapy in hepatocellular carcinoma , 2023, The Journal of clinical investigation.

[2]  Ruonan Zhang,et al.  TET2 suppresses VHL deficiency-driven clear cell renal cell carcinoma by inhibiting HIF signaling. , 2022, Cancer research.

[3]  D. Sabatini,et al.  mTOR at the nexus of nutrition, growth, ageing and disease , 2020, Nature Reviews Molecular Cell Biology.

[4]  Matthew D. Smith,et al.  Tumor suppressor TET2 promotes cancer immunity and immunotherapy efficacy. , 2019, The Journal of clinical investigation.

[5]  Dan Xie,et al.  5-methylcytosine promotes pathogenesis of bladder cancer through stabilizing mRNAs , 2019, Nature Cell Biology.

[6]  Le Li,et al.  p53 regulation of ammonia metabolism through urea cycle controls polyamine biosynthesis , 2019, Nature.

[7]  B. Manning,et al.  Molecular logic of mTORC1 signalling as a metabolic rheostat , 2019, Nature Metabolism.

[8]  K. Guan,et al.  mTOR as a central hub of nutrient signalling and cell growth , 2019, Nature Cell Biology.

[9]  Fei Lan,et al.  Glucose-regulated phosphorylation of TET2 by AMPK reveals a pathway linking diabetes to cancer , 2018, Nature.

[10]  Nan Li,et al.  Tet2 promotes pathogen infection-induced myelopoiesis through mRNA oxidation , 2018, Nature.

[11]  D. Sabatini Twenty-five years of mTOR: Uncovering the link from nutrients to growth , 2017, Proceedings of the National Academy of Sciences.

[12]  Yi Zhang,et al.  TET-mediated active DNA demethylation: mechanism, function and beyond , 2017, Nature Reviews Genetics.

[13]  D. Sabatini,et al.  mTOR Signaling in Growth, Metabolism, and Disease , 2017, Cell.

[14]  Matthew A. Cooper,et al.  Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice , 2017, Science.

[15]  Simone Sidoli,et al.  High-Resolution Mapping of RNA-Binding Regions in the Nuclear Proteome of Embryonic Stem Cells. , 2016, Molecular cell.

[16]  Bram Boeckx,et al.  Tumor hypoxia causes DNA hypermethylation by reducing TET activity , 2016, Nature.

[17]  Gregory A. Wyant,et al.  The CASTOR Proteins Are Arginine Sensors for the mTORC1 Pathway , 2016, Cell.

[18]  Xia Li,et al.  Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6 , 2015, Nature.

[19]  B. Porse,et al.  Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis , 2015, Genes & development.

[20]  Y. Xiong,et al.  WT1 recruits TET2 to regulate its target gene expression and suppress leukemia cell proliferation. , 2015, Molecular cell.

[21]  Xian Chen,et al.  CRL4(VprBP) E3 ligase promotes monoubiquitylation and chromatin binding of TET dioxygenases. , 2015, Molecular cell.

[22]  Gregory A. Wyant,et al.  Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1 , 2015, Science.

[23]  K. Guan,et al.  Differential regulation of mTORC1 by leucine and glutamine , 2015, Science.

[24]  T. van Dyke,et al.  Akt-dependent Activation of mTORC1 Complex Involves Phosphorylation of mTOR (Mammalian Target of Rapamycin) by IκB Kinase α (IKKα)* , 2014, The Journal of Biological Chemistry.

[25]  Z. Ling,et al.  Tumor development is associated with decrease of TET gene expression and 5-methylcytosine hydroxylation , 2013, Oncogene.

[26]  W. Reik,et al.  Nanog-dependent function of Tet1 and Tet2 in establishment of pluripotency , 2013, Nature.

[27]  N. Heintz,et al.  MeCP2 binds to 5hmc enriched within active genes and accessible chromatin in the nervous system , 2012, Cell.

[28]  J. Blenis,et al.  Hippo–YAP and mTOR pathways collaborate to regulate organ size , 2012, Nature Cell Biology.

[29]  Abraham J. Khorasani,et al.  Loss of 5-Hydroxymethylcytosine Is an Epigenetic Hallmark of Melanoma , 2012, Cell.

[30]  G. Bhagat,et al.  Early-stage epigenetic modification during somatic cell reprogramming by Parp1 and Tet2 , 2012, Nature.

[31]  Yong Chen,et al.  MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB , 2012, Autophagy.

[32]  Poshen B. Chen,et al.  Mbd3/NURD Complex Regulates Expression of 5-Hydroxymethylcytosine Marked Genes in Embryonic Stem Cells , 2011, Cell.

[33]  G. Schackert,et al.  5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations. , 2011, Cancer research.

[34]  Feng-Chun Yang,et al.  Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. , 2011, Blood.

[35]  Chuan He,et al.  Tet Proteins Can Convert 5-Methylcytosine to 5-Formylcytosine and 5-Carboxylcytosine , 2011, Science.

[36]  Yang Wang,et al.  Tet-Mediated Formation of 5-Carboxylcytosine and Its Excision by TDG in Mammalian DNA , 2011, Science.

[37]  K. Rajewsky,et al.  Ten-Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice , 2011, Proceedings of the National Academy of Sciences.

[38]  O. Abdel-Wahab,et al.  Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. , 2011, Cancer cell.

[39]  P. Opolon,et al.  TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. , 2011, Cancer cell.

[40]  Bin Wang,et al.  Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. , 2011, Cancer cell.

[41]  J. Licht,et al.  Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. , 2010, Cancer cell.

[42]  L. Aravind,et al.  Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2 , 2010, Nature.

[43]  J. Soulier,et al.  Mutation in TET2 in myeloid cancers. , 2009, The New England journal of medicine.

[44]  David R. Liu,et al.  Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1 , 2009 .

[45]  S. Baker,et al.  PTEN and the PI3-kinase pathway in cancer. , 2009, Annual review of pathology.

[46]  J. Bos,et al.  Regulation of the small GTPase Rheb by amino acids , 2006, Oncogene.