Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 415, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38580843

ABSTRACT

The ten-eleven-translocation family of proteins (TET1/2/3) are epigenetic regulators of gene expression. They regulate genes by promoting DNA demethylation (i.e., catalytic activity) and by partnering with regulatory proteins (i.e., non-catalytic functions). Unlike Tet1 and Tet2, Tet3 is not expressed in mouse embryonic stem cells (ESCs) but is induced upon ESC differentiation. However, the significance of its dual roles in lineage specification is less defined. By generating TET3 catalytic-mutant (Tet3m/m) and knockout (Tet3-/-) mouse ESCs and differentiating them to neuroectoderm (NE), we identify distinct catalytic-dependent and independent roles of TET3 in NE specification. We find that the catalytic activity of TET3 is important for activation of neural genes while its non-catalytic functions are involved in suppressing mesodermal programs. Interestingly, the vast majority of differentially methylated regions (DMRs) in Tet3m/m and Tet3-/- NE cells are hypomethylated. The hypo-DMRs are associated to aberrantly upregulated genes while the hyper-DMRs are linked to downregulated neural genes. We find the maintenance methyltransferase Dnmt1 as a direct target of TET3, which is downregulated in TET3-deficient NE cells and may contribute to the increased DNA hypomethylation. Our findings establish that the catalytic-dependent and -independent roles of TET3 have distinct contributions to NE specification with potential implications in development.


Subject(s)
Dioxygenases , Animals , Mice , Cell Differentiation/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Neural Plate/metabolism
2.
iScience ; 26(7): 107170, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37456851

ABSTRACT

Tet2 is a member of the Ten-eleven translocation (Tet1/2/3) family of enzymes and is expressed in embryonic stem cells (ESCs). It demethylates DNA (catalytic functions) and partners with chromatin modifiers (noncatalytic functions) to regulate genes. However, the significance of these functions in ESCs is less defined. Using Tet2 catalytic mutant (Tet2m/m) and knockout (Tet2-/-) ESCs, we identified Tet2 target genes regulated by its catalytic dependent versus independent roles. Tet2 was enriched at their active enhancers and promoters to demethylate them. We also identified the histone deacetylase component Sin3a as a Tet2 partner, co-localizing at promoters and active enhancers. Tet2 deficiency diminished Sin3a at these regions. Tet2 and Sin3a co-occupancy overlapped with Tet1. Combined loss of Tet1/2, but not of their catalytic activities, reduced Sin3a at active enhancers. These findings establish Tet2 catalytic and noncatalytic functions as regulators of DNA demethylation and Sin3a recruitment at active enhancers in ESCs.

3.
Exp Hematol ; 124: 45-55.e2, 2023 08.
Article in English | MEDLINE | ID: mdl-37225048

ABSTRACT

TET2 is a member of the Ten-eleven translocation (Tet) family of DNA dioxygenases that regulate gene expression by promoting DNA demethylation (enzymatic activity) and partnering with chromatin regulatory complexes (nonenzymatic functions). TET2 is highly expressed in the hematopoietic lineage, where its molecular functions are the subject of continuous investigations because of the prevalence of TET2 mutations in hematologic malignancies. Previously, we have implicated Tet2 catalytic and noncatalytic functions in the regulation of myeloid and lymphoid lineages, respectively. However, the impact of these functions of Tet2 on hematopoiesis as the bone marrow ages remains unclear. Here, we conducted comparative transplantations and transcriptomic analyses of 3-, 6-, 9-, and 12-month-old Tet2 catalytic mutant (Mut) and knockout (KO) bone marrow. Tet2 Mut bone marrow of all ages exclusively caused hematopoietic disorders of the myeloid lineage. In contrast, young Tet2 KO bone marrow developed both lymphoid and myeloid diseases, whereas older Tet2 KO bone marrow predominantly elicited myeloid disorders with shorter latency than age-matched Tet2 Mut bone marrow. We identified robust gene dysregulation in Tet2 KO Lin- cells at 6 months that involved lymphoma and myelodysplastic syndrome and/or leukemia-causing genes, many of which were hypermethylated early in life. There was a shift from lymphoid to myeloid gene deregulation in Tet2 KO Lin- cells with age, underpinning the higher incidence of myeloid diseases. These findings expand on the dynamic regulation of bone marrow by Tet2 and show that its catalytic-dependent and -independent roles have distinct impacts on myeloid and lymphoid lineages with age.


Subject(s)
Dioxygenases , Hematologic Diseases , Hematologic Neoplasms , Myelodysplastic Syndromes , Humans , Infant , Bone Marrow/metabolism , Hematologic Neoplasms/genetics , Myelodysplastic Syndromes/metabolism , Hematopoiesis/genetics , Hematologic Diseases/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Mutation
4.
Cells ; 11(8)2022 04 17.
Article in English | MEDLINE | ID: mdl-35456045

ABSTRACT

Ten eleven translocation 1 (Tet1) is a DNA dioxygenase that promotes DNA demethylation by oxidizing 5-methylcytosine. It can also partner with chromatin-activating and repressive complexes to regulate gene expressions independent of its enzymatic activity. Tet1 is highly expressed in embryonic stem cells (ESCs) and regulates pluripotency and differentiation. However, its roles in ESC cell cycle progression and proliferation have not been investigated. Using a series of Tet1 catalytic mutant (Tet1m/m), knockout (Tet1-/-) and wild type (Tet1+/+) mouse ESCs (mESCs), we identified a non-catalytic role of Tet1 in the proper cell cycle progression and proliferation of mESCs. Tet1-/-, but not Tet1m/m, mESCs exhibited a significant reduction in proliferation and delayed progression through G1. We found that the cyclin-dependent kinase inhibitor p21/Cdkn1a was uniquely upregulated in Tet1-/- mESCs and its knockdown corrected the slow proliferation and delayed G1 progression. Mechanistically, we found that p21 was a direct target of Tet1. Tet1 occupancy at the p21 promoter overlapped with the repressive histone mark H3K27me3 as well as with the H3K27 trimethyl transferase PRC2 component Ezh2. A loss of Tet1, but not loss of its catalytic activity, significantly reduced the enrichment of Ezh2 and H3K27 trimethylation at the p21 promoter without affecting the DNA methylation levels. We also found that the proliferation defects of Tet1-/- mESCs were independent of their differentiation defects. Together, these findings established a non-catalytic role for Tet1 in suppressing p21 in mESCs to ensure a rapid G1-to-S progression, which is a key hallmark of ESC proliferation. It also established Tet1 as an epigenetic regulator of ESC proliferation in addition to its previously defined roles in ESC pluripotency and differentiation.


Subject(s)
DNA-Binding Proteins , Proto-Oncogene Proteins , Animals , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Translocation, Genetic
5.
Stem Cell Res ; 61: 102770, 2022 05.
Article in English | MEDLINE | ID: mdl-35390758

ABSTRACT

The Inhibitor of disheveled and axin (Idax) and its ortholog the Retinoid inducible nuclear factor (Rinf) are DNA binding proteins with nuclear and cytoplasmic functions. Rinf is expressed in embryonic stem cells (ESCs) where it regulates transcription of the Ten-eleven translocation (Tet) enzymes, promoting neural and suppressing mesendoderm/trophectoderm differentiation. Here, we find that Idax, which is not expressed in ESCs, is induced upon differentiation. Like Rinf, Idax facilitates neural and silences trophectodermal programs. Individual or combined loss of Idax and Rinf led to downregulation of neural and upregulation of trophectoderm markers during differentiation of ESCs to embryoid bodies as well as during directed differentiation of ESCs to neural progenitor cells (NPCs) and trophoblast-like cells. These defects resemble those of Tet-deficient ESCs. Consistently, Tet genes are direct targets of Idax and Rinf, and loss of Idax and Rinf led to downregulation of Tet enzymes during ESC differentiation to NPCs and trophoblast-like cells. While Idax and Rinf single and double knockout (DKO) mice were viable and overtly normal, DKO embryos had reduced expression of several NPC markers in embryonic forebrains and deregulated expression of selected trophoblast markers in placentas. NPCs derived from DKO forebrains had reduced self-renewal while DKO placentas had increased junctional zone and reduced labyrinth layers. Together, our findings establish Idax and Rinf as regulators of Tet enzymes for proper differentiation of ESCs.


Subject(s)
DNA-Binding Proteins/metabolism , Neural Stem Cells , Animals , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Mice , Neural Stem Cells/metabolism
6.
Sci Adv ; 8(9): eabm3470, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35235365

ABSTRACT

Ten-eleven translocation (Tet) enzymes promote DNA demethylation by oxidizing 5-methylcytosine. They are expressed during development and are essential for mouse gastrulation. However, their postgastrulation functions are not well established. We find that global or endothelial-specific loss of all three Tet enzymes immediately after gastrulation leads to reduced number of hematopoietic stem and progenitor cells (HSPCs) and lethality in mid-gestation mouse embryos. This is due to defects in specification of HSPCs from endothelial cells (ECs) that compromise primitive and definitive hematopoiesis. Mechanistically, loss of Tet enzymes in ECs led to hypermethylation and down-regulation of NFκB1 and master hematopoietic transcription factors (Gata1/2, Runx1, and Gfi1b). Restoring Tet catalytic activity or overexpression of these factors in Tet-deficient ECs rescued hematopoiesis defects. This establishes Tet enzymes as activators of hematopoiesis programs in ECs for specification of HSPCs during embryogenesis, which is distinct from their roles in adult hematopoiesis, with implications in deriving HSPCs from pluripotent cells.


Subject(s)
Dioxygenases , Animals , Cell Differentiation/genetics , DNA Demethylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Embryonic Development/genetics , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Mammals/metabolism , Mice
7.
Nucleic Acids Res ; 50(6): 3169-3189, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35150568

ABSTRACT

Tet enzymes (Tet1/2/3) oxidize 5-methylcytosine to promote DNA demethylation and partner with chromatin modifiers to regulate gene expression. Tet1 is highly expressed in embryonic stem cells (ESCs), but its enzymatic and non-enzymatic roles in gene regulation are not dissected. We have generated Tet1 catalytically inactive (Tet1m/m) and knockout (Tet1-/-) ESCs and mice to study these functions. Loss of Tet1, but not loss of its catalytic activity, caused aberrant upregulation of bivalent (H3K4me3+; H3K27me3+) developmental genes, leading to defects in differentiation. Wild-type and catalytic-mutant Tet1 occupied similar genomic loci which overlapped with H3K27 tri-methyltransferase PRC2 and the deacetylase complex Sin3a at promoters of bivalent genes and with the helicase Chd4 at active genes. Loss of Tet1, but not loss of its catalytic activity, impaired enrichment of PRC2 and Sin3a at bivalent promoters leading to reduced H3K27 trimethylation and deacetylation, respectively, in absence of any changes in DNA methylation. Tet1-/-, but not Tet1m/m, embryos expressed higher levels of Gata6 and were developmentally delayed. Thus, the critical functions of Tet1 in ESCs and early development are mediated through its non-catalytic roles in regulating H3K27 modifications to silence developmental genes, and are more important than its catalytic functions in DNA demethylation.


Subject(s)
DNA-Binding Proteins , Dioxygenases , Embryonic Stem Cells , Proto-Oncogene Proteins , Animals , Cell Differentiation/genetics , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Embryonic Stem Cells/metabolism , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
8.
Dev Cell ; 56(22): 3052-3065.e5, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34710357

ABSTRACT

Loss of imprinting (LOI) results in severe developmental defects, but the mechanisms preventing LOI remain incompletely understood. Here, we dissect the functional components of the imprinting control region of the essential Dlk1-Dio3 locus (called IG-DMR) in pluripotent stem cells. We demonstrate that the IG-DMR consists of two antagonistic elements: a paternally methylated CpG island that prevents recruitment of TET dioxygenases and a maternally unmethylated non-canonical enhancer that ensures expression of the Gtl2 lncRNA by counteracting de novo DNA methyltransferases. Genetic or epigenetic editing of these elements leads to distinct LOI phenotypes with characteristic alternations of allele-specific gene expression, DNA methylation, and 3D chromatin topology. Although repression of the Gtl2 promoter results in dysregulated imprinting, the stability of LOI phenotypes depends on the IG-DMR, suggesting a functional hierarchy. These findings establish the IG-DMR as a bipartite control element that maintains imprinting by allele-specific restriction of the DNA (de)methylation machinery.


Subject(s)
Alleles , Calcium-Binding Proteins/genetics , DNA Methylation/genetics , Intercellular Signaling Peptides and Proteins/genetics , Animals , Chromosomes/genetics , Genomic Imprinting/genetics , Iodide Peroxidase/genetics , Mice , Promoter Regions, Genetic/genetics , RNA, Long Noncoding/genetics
11.
Front Cell Dev Biol ; 9: 645335, 2021.
Article in English | MEDLINE | ID: mdl-33681230

ABSTRACT

Studies of tissue-specific epigenomes have revealed 5-hydroxymethylcytosine (5hmC) to be a highly enriched and dynamic DNA modification in the metazoan nervous system, inspiring interest in the function of this epigenetic mark in neurodevelopment and brain function. 5hmC is generated by oxidation of 5-methylcytosine (5mC), a process catalyzed by the ten-eleven translocation (TET) enzymes. 5hmC serves not only as an intermediate in DNA demethylation but also as a stable epigenetic mark. Here, we review the known functions of 5hmC and TET enzymes in neural progenitor cell biology and embryonic and postnatal neurogenesis. We also discuss how TET enzymes and 5hmC regulate neuronal activity and brain function and highlight their implications in human neurodevelopmental and neurodegenerative disorders. Finally, we present outstanding questions in the field and envision new research directions into the roles of 5hmC and TET enzymes in neurodevelopment.

12.
Trends Cancer ; 7(7): 635-646, 2021 07.
Article in English | MEDLINE | ID: mdl-33468438

ABSTRACT

The mechanisms governing the methylome profile of tumor suppressors and oncogenes have expanded with the discovery of oxidized states of 5-methylcytosine (5mC). Ten-eleven translocation (TET) enzymes are a family of dioxygenases that iteratively catalyze 5mC oxidation and promote cytosine demethylation, thereby creating a dynamic global and local methylation landscape. While the catalytic function of TET enzymes during stem cell differentiation and development have been well studied, less is known about the multifaceted roles of TET enzymes during carcinogenesis. This review outlines several tiers of TET regulation and overviews how TET deregulation promotes a cancer phenotype. Defining the tissue-specific and context-dependent roles of TET enzymes will deepen our understanding of the epigenetic perturbations that promote or inhibit carcinogenesis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Mixed Function Oxygenases/metabolism , Neoplasms/genetics , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Clinical Trials as Topic , DNA Methylation/drug effects , DNA Methylation/immunology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Dioxygenases/antagonists & inhibitors , Dioxygenases/genetics , Drug Synergism , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Mutation , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Treatment Outcome
13.
Elife ; 82019 11 01.
Article in English | MEDLINE | ID: mdl-31663852

ABSTRACT

Even though pancreatic ductal adenocarcinoma (PDAC) is associated with fibrotic stroma, the molecular pathways regulating the formation of cancer associated fibroblasts (CAFs) are not well elucidated. An epigenomic analysis of patient-derived and de-novo generated CAFs demonstrated widespread loss of cytosine methylation that was associated with overexpression of various inflammatory transcripts including CXCR4. Co-culture of neoplastic cells with CAFs led to increased invasiveness that was abrogated by inhibition of CXCR4. Metabolite tracing revealed that lactate produced by neoplastic cells leads to increased production of alpha-ketoglutarate (aKG) within mesenchymal stem cells (MSCs). In turn, aKG mediated activation of the demethylase TET enzyme led to decreased cytosine methylation and increased hydroxymethylation during de novo differentiation of MSCs to CAF. Co-injection of neoplastic cells with TET-deficient MSCs inhibited tumor growth in vivo. Thus, in PDAC, a tumor-mediated lactate flux is associated with widespread epigenomic reprogramming that is seen during CAF formation.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Cellular Reprogramming/drug effects , Epigenesis, Genetic/drug effects , Lactic Acid/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cellular Reprogramming/genetics , DNA Methylation/drug effects , Humans , Ketoglutaric Acids/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Neoplasm Invasiveness , Receptors, CXCR4/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Transcriptome/genetics , Pancreatic Neoplasms
14.
Cell Rep ; 28(10): 2480-2490.e4, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31484061

ABSTRACT

The Ten-eleven translocation (TET) enzymes regulate gene expression by promoting DNA demethylation and partnering with chromatin modifiers. TET2, a member of this family, is frequently mutated in hematological disorders. The contributions of TET2 in hematopoiesis have been attributed to its DNA demethylase activity, and the significance of its nonenzymatic functions has remained undefined. To dissect the catalytic and non-catalytic requirements of Tet2, we engineered catalytically inactive Tet2 mutant mice and conducted comparative analyses of Tet2 mutant and Tet2 knockout animals. Tet2 knockout mice exhibited expansion of hematopoietic stem and progenitor cells (HSPCs) and developed myeloid and lymphoid disorders, while Tet2 mutant mice predominantly developed myeloid malignancies reminiscent of human myelodysplastic syndromes. HSPCs from Tet2 knockout mice exhibited distinct gene expression profiles, including downregulation of Gata2. Overexpression of Gata2 in Tet2 knockout bone marrow cells ameliorated disease phenotypes. Our results reveal the non-catalytic roles of TET2 in HSPC homeostasis.


Subject(s)
Biocatalysis , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Homeostasis , Proto-Oncogene Proteins/metabolism , Animals , Bone Marrow/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dioxygenases , Gene Expression Regulation , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Mice, Knockout , Mutation/genetics , Phenotype , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics
15.
Cell Rep ; 28(8): 1993-2003.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433977

ABSTRACT

The Retinoid inducible nuclear factor (Rinf), also known as CXXC5, is a nuclear protein, but its functions in the context of the chromatin are poorly defined. We find that in mouse embryonic stem cells (mESCs), Rinf binds to the chromatin and is enriched at promoters and enhancers of Tet1, Tet2, and pluripotency genes. The Rinf-bound regions show significant overlapping occupancy of pluripotency factors Nanog, Oct4, and Sox2, as well as Tet1 and Tet2. We found that Rinf forms a complex with Nanog, Oct4, Tet1, and Tet2 and facilitates their proper recruitment to regulatory regions of pluripotency and Tet genes in ESCs to positively regulate their transcription. Rinf deficiency in ESCs reduces expression of Rinf target genes, including several pluripotency factors and Tet enzymes, and causes aberrant differentiation. Together, our findings establish Rinf as a regulator of the pluripotency network genes and Tet enzymes in ESCs.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Chromatin/metabolism , DNA-Binding Proteins/deficiency , Dioxygenases , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Mice , Mice, SCID , Nanog Homeobox Protein/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Transcription Factors/deficiency , Transcription, Genetic
16.
Genome Res ; 27(11): 1830-1842, 2017 11.
Article in English | MEDLINE | ID: mdl-28986391

ABSTRACT

Transcriptional deregulation of oncogenic pathways is a hallmark of cancer and can be due to epigenetic alterations. 5-Hydroxymethylcytosine (5-hmC) is an epigenetic modification that has not been studied in pancreatic cancer. Genome-wide analysis of 5-hmC-enriched loci with hmC-seal was conducted in a cohort of low-passage pancreatic cancer cell lines, primary patient-derived xenografts, and pancreatic controls and revealed strikingly altered patterns in neoplastic tissues. Differentially hydroxymethylated regions preferentially affected known regulatory regions of the genome, specifically overlapping with known H3K4me1 enhancers. Furthermore, base pair resolution analysis of cytosine methylation and hydroxymethylation with oxidative bisulfite sequencing was conducted and correlated with chromatin accessibility by ATAC-seq and gene expression by RNA-seq in pancreatic cancer and control samples. 5-hmC was specifically enriched at open regions of chromatin, and gain of 5-hmC was correlated with up-regulation of the cognate transcripts, including many oncogenic pathways implicated in pancreatic neoplasia, such as MYC, KRAS, VEGFA, and BRD4 Specifically, BRD4 was overexpressed and acquired 5-hmC at enhancer regions in the majority of neoplastic samples. Functionally, acquisition of 5-hmC at BRD4 promoter was associated with increase in transcript expression in reporter assays and primary samples. Furthermore, blockade of BRD4 inhibited pancreatic cancer growth in vivo. In summary, redistribution of 5-hmC and preferential enrichment at oncogenic enhancers is a novel regulatory mechanism in human pancreatic cancer.


Subject(s)
5-Methylcytosine/analogs & derivatives , Pancreatic Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Sequence Analysis, RNA/methods , 5-Methylcytosine/metabolism , Animals , Cell Line, Tumor , Epigenesis, Genetic , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome-Wide Association Study , Histones/metabolism , Humans , Mice , Neoplasm Transplantation , Patient-Specific Modeling
17.
Neuropsychopharmacology ; 42(8): 1657-1669, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28074830

ABSTRACT

Depression is a leading cause of disease burden, yet current therapies fully treat <50% of affected individuals. Increasing evidence implicates epigenetic mechanisms in depression and antidepressant action. Here we examined a possible role for the DNA dioxygenase, ten-eleven translocation protein 1 (TET1), in depression-related behavioral abnormalities. We applied chronic social defeat stress, an ethologically validated mouse model of depression-like behaviors, and examined Tet1 expression changes in nucleus accumbens (NAc), a key brain reward region. We show decreased Tet1 expression in NAc in stress-susceptible mice only. Surprisingly, selective knockout of Tet1 in NAc neurons of adult mice produced antidepressant-like effects in several behavioral assays. To identify Tet1 targets that mediate these actions, we performed RNAseq on NAc after conditional deletion of Tet1 and found that immune-related genes are the most highly dysregulated. Moreover, many of these genes are also upregulated in the NAc of resilient mice after chronic social defeat stress. These findings reveal a novel role for TET1, an enzyme important for DNA hydroxymethylation, in the brain's reward circuitry in modulating stress responses in mice. We also identify a subset of genes that are regulated by TET1 in this circuitry. These findings provide new insight into the pathophysiology of depression, which can aid in future antidepressant drug discovery efforts.


Subject(s)
Anxiety/physiopathology , DNA-Binding Proteins/physiology , Depression/physiopathology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins/physiology , Stress, Psychological/physiopathology , Animals , Anxiety/genetics , Behavior, Animal , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Depression/genetics , Disease Models, Animal , Gene Expression/genetics , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Up-Regulation
18.
Cell Rep ; 15(8): 1809-21, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27184841

ABSTRACT

Ten-eleven translocation (Tet) family proteins convert 5-methylcytosine to 5-hydroxymethylcytosine. We show that mouse embryonic stem cells (ESCs) depleted of Tet1 and/or Tet2 by RNAi exhibit short telomeres and chromosomal instability, concomitant with reduced telomere recombination. Tet1 and Tet2 double-knockout ESCs also display short telomeres but to a lesser extent. Notably, Tet1/2/3 triple-knockout ESCs show heterogeneous telomere lengths and increased frequency of telomere loss and chromosomal fusion. Mechanistically, Tets depletion or deficiency increases Dnmt3b and decreases 5hmC levels, resulting in elevated methylation levels at sub-telomeres. Consistently, knockdown of Dnmt3b or addition of 2i (MAPK and GSK3ß inhibitors), which also inhibits Dnmt3b, reduces telomere shortening, partially rescuing Tet1/2 deficiency. Interestingly, Tet1/2 double or Tet1/2/3 triple knockout in ESCs consistently upregulates Zscan4, which may counteract telomere shortening. Together, Tet enzymes play important roles in telomere maintenance and chromosomal stability of ESCs by modulating sub-telomeric methylation levels.


Subject(s)
Chromosomal Instability/genetics , DNA-Binding Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Telomere/metabolism , Animals , Chromosomes, Mammalian/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Gene Deletion , Mice , Mice, Knockout , Recombination, Genetic/genetics , Telomere Shortening , DNA Methyltransferase 3B
19.
Mol Cell Biol ; 36(3): 452-61, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26598602

ABSTRACT

DNA methylation is a dynamic epigenetic modification with an important role in cell fate specification and reprogramming. The Ten eleven translocation (Tet) family of enzymes converts 5-methylcytosine to 5-hydroxymethylcytosine, which promotes passive DNA demethylation and functions as an intermediate in an active DNA demethylation process. Tet1/Tet2 double-knockout mice are characterized by developmental defects and epigenetic instability, suggesting a requirement for Tet-mediated DNA demethylation for the proper regulation of gene expression during differentiation. Here, we used whole-genome bisulfite and transcriptome sequencing to characterize the underlying mechanisms. Our results uncover the hypermethylation of DNA methylation canyons as the genomic key feature of Tet1/Tet2 double-knockout mouse embryonic fibroblasts. Canyon hypermethylation coincided with disturbed regulation of associated genes, suggesting a mechanistic explanation for the observed Tet-dependent differentiation defects. Based on these results, we propose an important regulatory role of Tet-dependent DNA demethylation for the maintenance of DNA methylation canyons, which prevents invasive DNA methylation and allows functional regulation of canyon-associated genes.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , Fibroblasts/cytology , Proto-Oncogene Proteins/genetics , Transcriptome , Adipogenesis , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/metabolism , Dioxygenases , Epigenesis, Genetic , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Proto-Oncogene Proteins/metabolism
20.
Cell Rep ; 13(8): 1692-704, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26586431

ABSTRACT

TET1/2/3 are methylcytosine dioxygenases that regulate cytosine hydroxymethylation. Tet1/2 are abundantly expressed in HSC/HPCs and are implicated in hematological malignancies. Tet2 deletion in mice causes myeloid malignancies, while Tet1-null mice develop B cell lymphoma after an extended period of latency. Interestingly, TET1/2 are often concomitantly downregulated in acute B-lymphocytic leukemia. Here, we investigated the overlapping and non-redundant functions of Tet1/2 using Tet1/2 double-knockout (DKO) mice. DKO and Tet2(-/-) HSC/HPCs show overlapping and unique 5 hmC and 5 mC profiles. DKO mice exhibit strikingly decreased incidence and delayed onset of myeloid malignancies in comparison to Tet2(-/-) mice and in contrast develop lethal B cell malignancies. Transcriptome analysis of DKO tumors reveals expression changes in many genes dysregulated in human B cell malignancies, including LMO2, BCL6, and MYC. These results highlight the critical roles of TET1/2 individually and together in the pathogenesis of hematological malignancies.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Hematologic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cytosine/metabolism , DNA Methylation/physiology , Dioxygenases/metabolism , Down-Regulation/physiology , Humans , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...