Combined deficiency of Tet1 and Tet2 causes epigenetic abnormalities but is compatible with postnatal development.

Tet enzymes (Tet1/2/3) convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in various embryonic and adult tissues. Mice mutant for either Tet1 or Tet2 are viable, raising the question of whether these enzymes have overlapping roles in development. Here we have generated Tet1 and Tet2 double-knockout (DKO) embryonic stem cells (ESCs) and mice. DKO ESCs remained pluripotent but were depleted of 5hmC and caused developmental defects in chimeric embryos. While a fraction of double-mutant embryos exhibited midgestation abnormalities with perinatal lethality, viable and overtly normal Tet1/Tet2-deficient mice were also obtained. DKO mice had reduced 5hmC and increased 5mC levels and abnormal methylation at various imprinted loci. Nevertheless, animals of both sexes were fertile, with females having smaller ovaries and reduced fertility. Our data show that loss of both enzymes is compatible with development but promotes hypermethylation and compromises imprinting. The data also suggest a significant contribution of Tet3 to hydroxylation of 5mC during development.

[1]  Rui Liu,et al.  Tet1 controls meiosis by regulating meiotic gene expression , 2012, Nature.

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

[3]  M. Surani,et al.  Genome-Wide Reprogramming in the Mouse Germ Line Entails the Base Excision Repair Pathway , 2010, Science.

[4]  T. Bestor,et al.  Methylation dynamics of imprinted genes in mouse germ cells. , 2002, Genomics.

[5]  D. Page,et al.  Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. , 2011, Cell stem cell.

[6]  Suhua Feng,et al.  5-Hydroxymethylcytosine is associated with enhancers and gene bodies in human embryonic stem cells , 2011, Genome Biology.

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

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

[9]  M. Surani,et al.  Parallel mechanisms of epigenetic reprogramming in the germline. , 2012, Trends in genetics : TIG.

[10]  Riitta Lahesmaa,et al.  Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. , 2011, Cell stem cell.

[11]  R. Jaenisch,et al.  Somatic cell nuclear transfer and derivation of embryonic stem cells in the mouse. , 2008, Methods.

[12]  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.

[13]  W. Reik,et al.  Genomic imprinting: parental influence on the genome , 2001, Nature Reviews Genetics.

[14]  James A. Cuff,et al.  A Bivalent Chromatin Structure Marks Key Developmental Genes in Embryonic Stem Cells , 2006, Cell.

[15]  W. Reik,et al.  Uncovering the role of 5-hydroxymethylcytosine in the epigenome , 2011, Nature Reviews Genetics.

[16]  X. Shirley Liu,et al.  Tet3 CXXC Domain and Dioxygenase Activity Cooperatively Regulate Key Genes for Xenopus Eye and Neural Development , 2012, Cell.

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

[18]  T. Bestor,et al.  The Colorful History of Active DNA Demethylation , 2008, Cell.

[19]  Yi Zhang,et al.  Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification , 2010, Nature.

[20]  W. Reik,et al.  5-Hydroxymethylcytosine in the mammalian zygote is linked with epigenetic reprogramming. , 2011, Nature communications.

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

[22]  Xiaochun Yu,et al.  TET2 promotes histone O-GlcNAcylation during gene transcription , 2012, Nature.

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

[24]  F. Tuorto,et al.  Hydroxylation of 5-methylcytosine by TET2 maintains the active state of the mammalian HOXA cluster , 2012, Nature Communications.

[25]  K. Helin,et al.  Explorer TET 1 and hydroxymethylcytosine in transcription and DNA methylation fidelity , 2012 .

[26]  Yi Zhang,et al.  Active DNA demethylation: many roads lead to Rome , 2010, Nature Reviews Molecular Cell Biology.

[27]  G. Fan,et al.  A sensitive mass spectrometry method for simultaneous quantification of DNA methylation and hydroxymethylation levels in biological samples. , 2011, Analytical biochemistry.

[28]  Philipp Kapranov,et al.  Genome-wide mapping of 5-hydroxymethylcytosine in embryonic stem cells , 2011, Nature.

[29]  T. Down,et al.  Germline DNA Demethylation Dynamics and Imprint Erasure Through 5-Hydroxymethylcytosine , 2013, Science.

[30]  N. Heintz,et al.  The Nuclear DNA Base 5-Hydroxymethylcytosine Is Present in Purkinje Neurons and the Brain , 2009, Science.

[31]  K. Helin,et al.  DNA methylation: TET proteins—guardians of CpG islands? , 2012, EMBO reports.

[32]  K. Rajewsky,et al.  Tet2 Negatively Regulates Homeostasis and Differentiation of Hematopoietic Stem Cells in Mice , 2011 .

[33]  Keji Zhao,et al.  Dual functions of Tet1 in transcriptional regulation in mouse embryonic stem cells , 2011, Nature.

[34]  W. Reik,et al.  The Dynamics of Genome-wide DNA Methylation Reprogramming in Mouse Primordial Germ Cells , 2012, Molecular cell.

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

[36]  Z. Deng,et al.  The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes , 2011, Nature.

[37]  W. Reik,et al.  Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation , 2011, Nature.