Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biosci Rep ; 41(11)2021 11 26.
Article in English | MEDLINE | ID: mdl-34724040

ABSTRACT

Lysine methyltransferase 2D (KMT2D), as one of the key histone methyltransferases responsible for histone 3 lysine 4 methylation (H3K4me), has been proved to be the main pathogenic gene of Kabuki syndrome disease. Kabuki patients with KMT2D mutation frequently present various dental abnormalities, including abnormal tooth number and crown morphology. However, the exact function of KMT2D in tooth development remains unclear. In this report, we systematically elucidate the expression pattern of KMT2D in early tooth development and outline the molecular mechanism of KMT2D in dental epithelial cell line. KMT2D and H3K4me mainly expressed in enamel organ and Kmt2d knockdown led to the reduction in cell proliferation activity and cell cycling activity in dental epithelial cell line (LS8). RNA-sequencing (RNA-seq) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis screened out several important pathways affected by Kmt2d knockdown including Wnt signaling. Consistently, Top/Fop assay confirmed the reduction in Wnt signaling activity in Kmt2d knockdown cells. Nuclear translocation of ß-catenin was significantly reduced by Kmt2d knockdown, while lithium chloride (LiCl) partially reversed this phenomenon. Moreover, LiCl partially reversed the decrease in cell proliferation activity and G1 arrest, and the down-regulation of Wnt-related genes in Kmt2d knockdown cells. In summary, the present study uncovered a pivotal role of histone methyltransferase KMT2D in dental epithelium proliferation and cell cycle homeostasis partially through regulating Wnt/ß-catenin signaling. The findings are important for understanding the role of KMT2D and histone methylation in tooth development.


Subject(s)
Epithelial Cells/metabolism , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Tooth/metabolism , Wnt Signaling Pathway/genetics , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle/genetics , Cell Line , Cell Proliferation/genetics , Cyclin D1/metabolism , Epithelial Cells/cytology , Histones/metabolism , Lithium Chloride/pharmacology , Mice , Mice, Inbred ICR , Molar/metabolism , Tooth/cytology , Wnt Signaling Pathway/drug effects
2.
Commun Biol ; 3(1): 278, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32483278

ABSTRACT

Histone H3 lysine 4 methylation (H3K4me) is extensively regulated by numerous writer and eraser enzymes in mammals. Nine H3K4me enzymes are associated with neurodevelopmental disorders to date, indicating their important roles in the brain. However, interplay among H3K4me enzymes during brain development remains largely unknown. Here, we show functional interactions of a writer-eraser duo, KMT2A and KDM5C, which are responsible for Wiedemann-Steiner Syndrome (WDSTS), and mental retardation X-linked syndromic Claes-Jensen type (MRXSCJ), respectively. Despite opposite enzymatic activities, the two mouse models deficient for either Kmt2a or Kdm5c shared reduced dendritic spines and increased aggression. Double mutation of Kmt2a and Kdm5c clearly reversed dendritic morphology, key behavioral traits including aggression, and partially corrected altered transcriptomes and H3K4me landscapes. Thus, our study uncovers common yet mutually suppressive aspects of the WDSTS and MRXSCJ models and provides a proof of principle for balancing a single writer-eraser pair to ameliorate their associated disorders.


Subject(s)
Abnormalities, Multiple/genetics , Aggression , Craniofacial Abnormalities/genetics , Dendritic Spines/metabolism , Growth Disorders/genetics , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Hypertrichosis/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Disease Models, Animal , Histone Demethylases/deficiency , Histone-Lysine N-Methyltransferase/deficiency , Male , Methylation , Mice , Myeloid-Lymphoid Leukemia Protein/deficiency
3.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31557133

ABSTRACT

Kabuki syndrome 1 (KS1) is a Mendelian disorder of the epigenetic machinery caused by mutations in the gene encoding KMT2D, which methylates lysine 4 on histone H3 (H3K4). KS1 is characterized by intellectual disability, postnatal growth retardation, and distinct craniofacial dysmorphisms. A mouse model (Kmt2d+/ßGeo) exhibits features of the human disorder and has provided insight into other phenotypes; however, the mechanistic basis of skeletal abnormalities and growth retardation remains elusive. Using high-resolution micro-CT, we show that Kmt2d+/ßGeo mice have shortened long bones and ventral bowing of skulls. In vivo expansion of growth plates within skulls and long bones suggests disrupted endochondral ossification as a common disease mechanism. Stable chondrocyte cell lines harboring inactivating mutations in Kmt2d exhibit precocious differentiation, further supporting this mechanism. A known inducer of chondrogenesis, SOX9, and its targets show markedly increased expression in Kmt2d-/- chondrocytes. By transcriptome profiling, we identify Shox2 as a putative KMT2D target. We propose that decreased KMT2D-mediated H3K4me3 at Shox2 releases Sox9 inhibition and thereby leads to enhanced chondrogenesis, providing a potentially novel and plausible explanation for precocious chondrocyte differentiation. Our findings provide insight into the pathogenesis of growth retardation in KS1 and suggest therapeutic approaches for this and related disorders.


Subject(s)
Abnormalities, Multiple/genetics , Cell Differentiation/genetics , Chondrogenesis/genetics , Face/abnormalities , Hematologic Diseases/genetics , Histone-Lysine N-Methyltransferase/deficiency , Homeodomain Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/deficiency , Skull/growth & development , Vestibular Diseases/genetics , Abnormalities, Multiple/pathology , Animals , Chondrocytes/pathology , Disease Models, Animal , Face/pathology , Female , Hematologic Diseases/pathology , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Male , Mice , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Oxygen/metabolism , SOX9 Transcription Factor/metabolism , Skull/cytology , Skull/diagnostic imaging , Vestibular Diseases/pathology , X-Ray Microtomography
4.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31465303

ABSTRACT

Chromatin modifiers act to coordinate gene expression changes critical to neuronal differentiation from neural stem/progenitor cells (NSPCs). Lysine-specific methyltransferase 2D (KMT2D) encodes a histone methyltransferase that promotes transcriptional activation and is frequently mutated in cancers and in the majority (>70%) of patients diagnosed with the congenital, multisystem intellectual disability disorder Kabuki syndrome 1 (KS1). Critical roles for KMT2D are established in various non-neural tissues, but the effects of KMT2D loss in brain cell development have not been described. We conducted parallel studies of proliferation, differentiation, transcription, and chromatin profiling in KMT2D-deficient human and mouse models to define KMT2D-regulated functions in neurodevelopmental contexts, including adult-born hippocampal NSPCs in vivo and in vitro. We report cell-autonomous defects in proliferation, cell cycle, and survival, accompanied by early NSPC maturation in several KMT2D-deficient model systems. Transcriptional suppression in KMT2D-deficient cells indicated strong perturbation of hypoxia-responsive metabolism pathways. Functional experiments confirmed abnormalities of cellular hypoxia responses in KMT2D-deficient neural cells and accelerated NSPC maturation in vivo. Together, our findings support a model in which loss of KMT2D function suppresses expression of oxygen-responsive gene programs important to neural progenitor maintenance, resulting in precocious neuronal differentiation in a mouse model of KS1.


Subject(s)
Abnormalities, Multiple/genetics , Brain/growth & development , Cell Differentiation/genetics , DNA-Binding Proteins/deficiency , Face/abnormalities , Hematologic Diseases/genetics , Histone-Lysine N-Methyltransferase/deficiency , Myeloid-Lymphoid Leukemia Protein/deficiency , Neoplasm Proteins/deficiency , Neural Stem Cells/pathology , Neurons/pathology , Vestibular Diseases/genetics , Abnormalities, Multiple/pathology , Animals , Brain/cytology , Cell Hypoxia/genetics , Cell Proliferation/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Face/pathology , Female , Fibroblasts , Hematologic Diseases/pathology , Histone-Lysine N-Methyltransferase/genetics , Humans , Induced Pluripotent Stem Cells , Male , Mice , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Oxygen/metabolism , Primary Cell Culture , RNA-Seq , Single-Cell Analysis , Skin/cytology , Skin/pathology , Vestibular Diseases/pathology
5.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29916805

ABSTRACT

Hematopoietic stem cells require MLL1, which is one of six Set1/Trithorax-type histone 3 lysine 4 (H3K4) methyltransferases in mammals and clinically the most important leukemia gene. Here, we add to emerging evidence that all six H3K4 methyltransferases play essential roles in the hematopoietic system by showing that conditional mutagenesis of Setd1b in adult mice provoked aberrant homeostasis of hematopoietic stem and progenitor cells (HSPCs). Using both ubiquitous and hematopoietic-specific deletion strategies, the loss of Setd1b resulted in peripheral thrombo- and lymphocytopenia, multilineage dysplasia, myeloid-biased extramedullary hematopoiesis in the spleen, and lethality. By transplantation experiments and expression profiling, we determined that Setd1b is autonomously required in the hematopoietic lineages where it regulates key lineage specification components, including Cebpa, Gata1, and Klf1. Altogether, these data imply that the Set1/Trithorax-type epigenetic machinery sustains different aspects of hematopoiesis and constitutes a second framework additional to the transcription factor hierarchy of hematopoietic homeostasis.


Subject(s)
Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/genetics , Homeostasis/genetics , Lymphopenia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Thrombocytopenia/genetics , Animals , Bone Marrow Transplantation , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Lineage/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genes, Lethal , Hematopoietic Stem Cells/cytology , Histone-Lysine N-Methyltransferase/deficiency , Isoenzymes/deficiency , Isoenzymes/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lymphopenia/metabolism , Lymphopenia/pathology , Mice , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/deficiency , Spleen/metabolism , Spleen/pathology , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Whole-Body Irradiation
6.
Leukemia ; 31(10): 2200-2210, 2017 10.
Article in English | MEDLINE | ID: mdl-28210005

ABSTRACT

Rearrangements involving the NUP98 gene resulting in fusions to several partner genes occur in acute myeloid leukemia and myelodysplastic syndromes. This study demonstrates that the second FG repeat domain of the NUP98 moiety of the NUP98-HOXA9 fusion protein is important for its cell immortalization and leukemogenesis activities. We demonstrate that NUP98-HOXA9 interacts with mixed lineage leukemia (MLL) via this FG repeat domain and that, in the absence of MLL, NUP98-HOXA9-induced cell immortalization and leukemogenesis are severely inhibited. Molecular analyses indicate that MLL is important for the recruitment of NUP98-HOXA9 to the HOXA locus and for NUP98-HOXA9-induced HOXA gene expression. Our data indicate that MLL is crucial for NUP98-HOXA9 leukemia initiation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Leukemic/genetics , Histone-Lysine N-Methyltransferase/physiology , Homeodomain Proteins/physiology , Leukemia, Experimental/genetics , Myeloid-Lymphoid Leukemia Protein/physiology , Nuclear Pore Complex Proteins/physiology , Oncogene Proteins, Fusion/physiology , Animals , Chromatin Immunoprecipitation , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Leukemia, Experimental/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Protein Binding , Protein Domains , Protein Interaction Mapping , Radiation Chimera , Transfection
7.
Neuropsychopharmacology ; 41(13): 3103-3113, 2016 12.
Article in English | MEDLINE | ID: mdl-27485686

ABSTRACT

Lysine (K) methyltransferase 2a (Kmt2a) and other regulators of H3 lysine 4 methylation, a histone modification enriched at promoters and enhancers, are widely expressed throughout the brain, but molecular and cellular phenotypes in subcortical areas remain poorly explored. We report that Kmt2a conditional deletion in postnatal forebrain is associated with excessive nocturnal activity and with absent or blunted responses to stimulant and dopaminergic agonist drugs, in conjunction with near-complete loss of spike-timing-dependent long-term potentiation in medium spiny neurons (MSNs). Selective ablation of Kmt2a, but not the ortholog Kmt2b, in adult ventral striatum/nucleus accumbens neurons markedly increased anxiety scores in multiple behavioral paradigms. Striatal transcriptome sequencing in adult mutants identified 262 Kmt2a-sensitive genes, mostly downregulated in Kmt2a-deficient mice. Transcriptional repression includes the 5-Htr2a serotonin receptor, strongly associated with anxiety- and depression-related disorders in human and animal models. Consistent with the role of Kmt2a in promoting gene expression, the transcriptional regulators Bahcc1, Isl1, and Sp9 were downregulated and affected by H3K4 promoter hypomethylation. Therefore, Kmt2a regulates synaptic plasticity in striatal neurons and provides an epigenetic drug target for anxiety and dopamine-mediated behaviors.


Subject(s)
Action Potentials/genetics , Anxiety , Dopamine Agents/pharmacology , Histone-Lysine N-Methyltransferase/deficiency , Myeloid-Lymphoid Leukemia Protein/deficiency , Neuronal Plasticity/genetics , Neurons/physiology , Ventral Striatum/cytology , Action Potentials/drug effects , Animals , Animals, Newborn , Anxiety/drug therapy , Anxiety/genetics , Anxiety/metabolism , Anxiety/physiopathology , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Disease Models, Animal , Female , Histone-Lysine N-Methyltransferase/genetics , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oligonucleotide Array Sequence Analysis , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Cell Stem Cell ; 18(4): 481-94, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26996599

ABSTRACT

The interconversion between naive and primed pluripotent states is accompanied by drastic epigenetic rearrangements. However, it is unclear whether intrinsic epigenetic events can drive reprogramming to naive pluripotency or if distinct chromatin states are instead simply a reflection of discrete pluripotent states. Here, we show that blocking histone H3K4 methyltransferase MLL1 activity with the small-molecule inhibitor MM-401 reprograms mouse epiblast stem cells (EpiSCs) to naive pluripotency. This reversion is highly efficient and synchronized, with more than 50% of treated EpiSCs exhibiting features of naive embryonic stem cells (ESCs) within 3 days. Reverted ESCs reactivate the silenced X chromosome and contribute to embryos following blastocyst injection, generating germline-competent chimeras. Importantly, blocking MLL1 leads to global redistribution of H3K4me1 at enhancers and represses lineage determinant factors and EpiSC markers, which indirectly regulate ESC transcription circuitry. These findings show that discrete perturbation of H3K4 methylation is sufficient to drive reprogramming to naive pluripotency.


Subject(s)
Cellular Reprogramming/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Mouse Embryonic Stem Cells/drug effects , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Oligopeptides/pharmacology , Pluripotent Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Animals , Cell Line , Germ Layers/drug effects , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/metabolism , Pluripotent Stem Cells/metabolism
9.
BMC Med Genomics ; 8: 74, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26553048

ABSTRACT

BACKGROUND: TGFß1-induced expression of transforming growth factor ß-induced protein (TGFBIp) and extracellular matrix (ECM) genes plays a major role in the development of granular corneal dystrophy type 2 (GCD2: also called Avellino corneal dystrophy). Although some key transcription factors are known, the epigenetic mechanisms modulating TGFBIp and ECM expression remain unclear. We examined the role of chromatin markers such as histone H3 lysine methylation (H3Kme) in TGFß1-induced TGFBIp and ECM gene expression in normal and GCD2-derived human corneal fibroblasts. METHODS: Wild-type (n = 3), GCD2-heterozygous (n = 1), and GCD2-homozygous (n = 3) primary human corneal fibroblasts were harvested from human donors and patients prepared. Microarray and gene-expression profiling, Chromatin immunoprecipitation microarray analysis, and Methylated DNA isolation assay-assisted CpG microarrays was performed in Wild-type and GCD2-homozygous human cells. RESULTS: Transcription and extracellular-secretion levels of TGFBIp were high in normal cells compared with those in GCD2-derived cells and were related to H3K4me3 levels but not to DNA methylation over the TGFBI locus. TGFß1 increased the expression of TGFBIp and the ECM-associated genes connective tissue growth factor, collagen-α2[Ι], and plasminogen activator inhibitor-1 in normal corneal fibroblasts. Increased levels of gene-activating markers (H3K4me1/3) and decreased levels of repressive markers (H3K27me3) at the promoters of those gene accompanied the changes in expression. TGFß1 also increased recruitment of the H3K4 methyltransferase MLL1 and of SET7/9 and also the binding of Smad3 to the promoters. Knockdown of both MLL1 and SET7/9 significantly blocked the TGFß1-induced gene expression and inhibited TGFß1-induced changes in promoter H3K4me1/3 levels. Those effects were very weak, however, in GCD2-derived corneal fibroblasts. CONCLUSIONS: Taken together, the results show the functional role of H3K4me in TGFß1-mediated TGFBIp and ECM gene expression in corneal fibroblasts. Pharmacologic and other therapies that regulate these modifications could have potential cornea-protective effects for granular corneal dystrophy.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/pathology , Gene Expression Regulation , Histones/metabolism , Transforming Growth Factor beta/genetics , Cornea/cytology , Cornea/pathology , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Extracellular Matrix Proteins/metabolism , Gene Knockdown Techniques , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Homozygote , Humans , Lysine/metabolism , Methylation , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Transport , Smad3 Protein/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism
10.
J Clin Invest ; 125(5): 2007-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25866973

ABSTRACT

Rapidly cycling fetal and neonatal hematopoietic stem cells (HSCs) generate a pool of quiescent adult HSCs after establishing hematopoiesis in the bone marrow. We report an essential role for the trithorax group gene absent, small, or homeotic 1-like (Ash1l) at this developmental transition. Emergence and expansion of Ash1l-deficient fetal/neonatal HSCs were preserved; however, in young adult animals, HSCs were profoundly depleted. Ash1l-deficient adult HSCs had markedly decreased quiescence and reduced cyclin-dependent kinase inhibitor 1b/c (Cdkn1b/1c) expression and failed to establish long-term trilineage bone marrow hematopoiesis after transplantation to irradiated recipients. Wild-type HSCs could efficiently engraft when transferred to unirradiated, Ash1l-deficient recipients, indicating increased availability of functional HSC niches in these mice. Ash1l deficiency also decreased expression of multiple Hox genes in hematopoietic progenitors. Ash1l cooperated functionally with mixed-lineage leukemia 1 (Mll1), as combined loss of Ash1l and Mll1, but not isolated Ash1l or Mll1 deficiency, induced overt hematopoietic failure. Our results uncover a trithorax group gene network that controls quiescence, niche occupancy, and self-renewal potential in adult HSCs.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Histone-Lysine N-Methyltransferase/physiology , Anemia, Aplastic , Animals , Animals, Newborn , Bone Marrow Diseases , Bone Marrow Failure Disorders , Bone Marrow Transplantation , Cell Cycle/genetics , Cell Division/genetics , Colony-Forming Units Assay , DNA-Binding Proteins , Fluorouracil/toxicity , Gene Expression Regulation, Developmental , Graft Survival , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/pathology , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Liver/cytology , Liver/embryology , Liver/metabolism , Liver Transplantation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/physiology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Radiation Chimera , Stem Cell Niche
11.
Neurosurgery ; 75(4): 472-82; discussion 482, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24887289

ABSTRACT

BACKGROUND: Mixed lineage leukemia-1 (Mll1) epigenetically regulates gene expression patterns that specify cellular identity in both embryonic development and adult stem cell populations. In the adult mouse brain, multipotent neural stem cells (NSCs) in the subventricular zone generate new neurons throughout life, and Mll1 is required for this postnatal neurogenesis but not for glial cell differentiation. Analysis of Mll1-dependent transcription may identify neurogenic genes useful for the direct reprogramming of astrocytes into neurons. OBJECTIVE: To identify Mll1-dependent transcriptional modules and to determine whether genes in the neurogenic modules can be used to directly reprogram astrocytes into neurons. METHODS: We performed gene coexpression module analysis on microarray data from differentiating wild-type and Mll1-deleted subventricular zone NSCs. Key developmental regulators belonging to the neurogenic modules were overexpressed in Mll1-deleted cells and cultured cortical astrocytes, and cell phenotypes were analyzed by immunocytochemistry and electrophysiology. RESULTS: Transcriptional modules that correspond to neurogenesis were identified in wild-type NSCs. Modules related to astrocytes and oligodendrocytes were enriched in Mll1-deleted NSCs, consistent with their gliogenic potential. Overexpression of genes selected from the neurogenic modules enhanced the production of neurons from Mll1-deleted cells, and overexpression of Brn4 (Pou3f4) in nonneurogenic cortical astroglia induced their transdifferentiation into electrophysiologically active neurons. CONCLUSION: Our results demonstrate that Mll1 is required for the expression of neurogenic but not gliogenic transcriptional modules in a multipotent NSC population and further indicate that specific Mll1-dependent genes may be useful for direct reprogramming strategies.


Subject(s)
Astrocytes/physiology , Cell Transdifferentiation/physiology , Histone-Lysine N-Methyltransferase/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Nerve Tissue Proteins/physiology , Neural Stem Cells/physiology , Neurons/physiology , POU Domain Factors/physiology , Animals , Histone-Lysine N-Methyltransferase/deficiency , Mice , Microarray Analysis , Myeloid-Lymphoid Leukemia Protein/deficiency , Neurogenesis/physiology
12.
J Biol Chem ; 289(27): 18914-27, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24831003

ABSTRACT

Heat shock protein 90 (HSP90) inhibition inhibits cancer cell proliferation through depleting client oncoproteins and shutting down multiple oncogenic pathways. Therefore, it is an attractive strategy for targeting human cancers. Several HSP90 inhibitors, including AUY922 and STA9090, show promising effects in clinical trials. However, the efficacy of HSP90 inhibitors may be limited by heat shock factor 1 (HSF1)-mediated feedback mechanisms. Here, we identify, through an siRNA screen, that the histone H3 lysine 4 methyltransferase MLL1 functions as a coactivator of HSF1 in response to HSP90 inhibition. MLL1 is recruited to the promoters of HSF1 target genes and regulates their expression in response to HSP90 inhibition. In addition, a striking combination effect is observed when MLL1 depletion is combined with HSP90 inhibition in various human cancer cell lines and tumor models. Thus, targeting MLL1 may block a HSF1-mediated feedback mechanism induced by HSP90 inhibition and provide a new avenue to enhance HSP90 inhibitor activity in human cancers.


Subject(s)
DNA-Binding Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Transcription Factors/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Heat Shock Transcription Factors , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Humans , Isoxazoles/pharmacology , Mice , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Resorcinols/pharmacology , Transcription Factors/genetics
13.
Development ; 141(3): 526-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24423662

ABSTRACT

Trimethylation of histone H3 lysine 4 (H3K4me3) at the promoters of actively transcribed genes is a universal epigenetic mark and a key product of Trithorax group action. Here, we show that Mll2, one of the six Set1/Trithorax-type H3K4 methyltransferases in mammals, is required for trimethylation of bivalent promoters in mouse embryonic stem cells. Mll2 is bound to bivalent promoters but also to most active promoters, which do not require Mll2 for H3K4me3 or mRNA expression. By contrast, the Set1 complex (Set1C) subunit Cxxc1 is primarily bound to active but not bivalent promoters. This indicates that bivalent promoters rely on Mll2 for H3K4me3 whereas active promoters have more than one bound H3K4 methyltransferase, including Set1C. Removal of Mll1, sister to Mll2, had almost no effect on any promoter unless Mll2 was also removed, indicating functional backup between these enzymes. Except for a subset, loss of H3K4me3 on bivalent promoters did not prevent responsiveness to retinoic acid, thereby arguing against a priming model for bivalency. In contrast, we propose that Mll2 is the pioneer trimethyltransferase for promoter definition in the naïve epigenome and that Polycomb group action on bivalent promoters blocks the premature establishment of active, Set1C-bound, promoters.


Subject(s)
Embryonic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Promoter Regions, Genetic , Animals , Binding Sites/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chromosomes, Artificial, Bacterial/metabolism , Embryonic Stem Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Methylation/drug effects , Mice , Models, Biological , Myeloid-Lymphoid Leukemia Protein/deficiency , Protein Binding/drug effects , Protein Binding/genetics , Transgenes/genetics , Tretinoin/pharmacology
14.
Cell Cycle ; 12(18): 2969-72, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23974107

ABSTRACT

Mixed lineage leukemia 1 (MLL1) is a gene that is disrupted by chromosomal translocation characteristically in a large proportion of infant leukemia and also in a fraction of childhood and adult leukemia. MLL1 encodes a chromatin regulatory protein related to the Drosophila Trithorax protein, a well-studied epigenetic factor that functions during development to maintain expression of its target genes. Although tremendous progress has been made understanding the downstream targets of MLL1 fusion oncoproteins and how manipulation of those targets impacts leukemogenesis, very little is known regarding how the initial expression of an MLL1 fusion protein impacts on that cell's behavior, particularly how the cell cycle is affected. Here, we focused on the function of endogenous MLL1 in the stem and progenitor cell types that are likely to be transformed upon MLL1 translocation. Our studies reveal a differential response of stem or progenitor populations to acute loss of MLL1 on proliferation and survival. These data suggest that the effects of MLL1 fusion oncoproteins will initiate the leukemogenic process differentially depending on the differentiation state of the cell type in which the translocation occurs.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CD48 Antigen , Cell Proliferation , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/genetics , Leukemia/metabolism , Leukemia/pathology , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism
15.
Oncogene ; 32(28): 3359-70, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-22926525

ABSTRACT

Mixed lineage leukemias (MLLs) are human histone H3 lysine-4-specific methyl transferases that have critical roles in gene expression, epigenetics and cancer. Herein, we demonstrated that antisense-mediated knockdown of MLL1 induced cell-cycle arrest and apoptosis in cultured cells. Intriguingly, application of MLL1 antisense specifically knocked down MLL1 in vivo and suppressed the growth of xenografted cervical tumor implanted in nude mouse. MLL1 knockdown downregulated various growth and angiogenic factors, such as HIF1α, VEGF and CD31, in tumor tissue affecting tumor growth. MLL1 is overexpressed along the line of vascular network and localized adjacent to endothelial cell layer expressing CD31, indicating potential roles of MLL1 in vasculogenesis. MLL1 is also overexpressed in the hypoxic regions along with HIF1α. Overall, our studies demonstrated that MLL1 is a key factor in hypoxia signaling, vasculogenesis and tumor growth, and its depletion suppresses tumor growth in vivo, indicating its potential in novel cancer therapy.


Subject(s)
Gene Knockdown Techniques , Myeloid-Lymphoid Leukemia Protein/metabolism , Neovascularization, Pathologic/enzymology , Uterine Cervical Neoplasms/blood supply , Uterine Cervical Neoplasms/pathology , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Transformation, Neoplastic , Female , Histone-Lysine N-Methyltransferase , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Signal Transduction/genetics , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics
16.
J Cell Sci ; 125(Pt 17): 4058-66, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22623725

ABSTRACT

Genes of the mixed lineage leukemia (MLL) family regulate transcription by methylating histone H3K4. Six members of the MLL family exist in humans, including SETD1A, SETD1B and MLL1-MLL4. Each of them plays non-redundant roles in development and disease genesis. MLL1 regulates the cell cycle and the oscillation of circadian gene expression. Its fusion proteins are involved in leukemogenesis. Here, we studied the role of MLL1 in innate immunity and found it selectively regulates the activation of genes downstream of NF-κB mediated by tumor necrosis factor (TNFα) and lipopolysaccharide (LPS). Real-time PCR and genome-wide gene expression profile analysis proved that the deficiency of MLL1 reduced the expression of a group of genes downstream of nuclear factor κB (NF-κB). However, the activation of NF-κB itself was not affected. The MLL1 complex is found both in the nucleus and cytoplasm and is associated with NF-κB. CHIP assays proved that the translocation of MLL1 to chromatin was dependent on NF-κB. Our results suggest that MLL1 is recruited to its target genes by activated NF-κB and regulates their transcription.


Subject(s)
Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins , Embryo, Mammalian/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Genome/genetics , HEK293 Cells , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Myeloid-Lymphoid Leukemia Protein/deficiency , NF-KappaB Inhibitor alpha , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Transport/drug effects , Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Superoxide Dismutase/metabolism , Transcription Factor RelA/metabolism
17.
PLoS One ; 6(10): e25449, 2011.
Article in English | MEDLINE | ID: mdl-21998658

ABSTRACT

Translocations involving the Mixed Lineage Leukemia (MLL) gene generate in-frame fusions of MLL with more than 50 different partner genes (PGs). Common to all MLL translocations is the exchange not only of coding regions, but also of MLL and PG 3'-untranslated regions (3'UTRs). As a result, the MLL-PG fusion is normally highly expressed and considered the main driver of leukemia development, whereas the function of the PG-MLL fusions in leukemic disease is unclear. As 3'UTRs have been recognized as determinant regions for regulation of gene expression, we hypothesized that loss of the MLL 3'UTR could have a role in generating high MLL-PG levels and leukemia development. Here, we first tested the MLL-PG and PG-MLL mRNA levels in different leukemic cells and tumours and uncovered differential expression that indicates strong repression by the MLL-3'UTR. Reporter assays confirmed that the 3'UTR of MLL, but not of its main PGs, harbours a region that imposes a strong gene silencing effect. Gene suppression by the MLL 3'UTR was largely microRNA independent and did not affect mRNA stability, but inhibited transcription. This effect can at least partially be attributed to a tighter interaction of the MLL 3'UTR with RNA polymerase II than PG 3'UTRs, affecting its phosphorylation state. Altogether, our findings indicate that MLL translocations relieve oncogenic MLL-PG fusions from the repressive MLL 3'UTR, contributing to higher activity of these genes and leukaemia development.


Subject(s)
3' Untranslated Regions/genetics , Gene Silencing , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Fusion/genetics , Histone-Lysine N-Methyltransferase , Humans , Leukemia/genetics , Leukemia/pathology , Male , Translocation, Genetic/genetics
18.
Nature ; 467(7313): 343-6, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20818375

ABSTRACT

Cell cycle checkpoints are implemented to safeguard the genome, avoiding the accumulation of genetic errors. Checkpoint loss results in genomic instability and contributes to the evolution of cancer. Among G1-, S-, G2- and M-phase checkpoints, genetic studies indicate the role of an intact S-phase checkpoint in maintaining genome integrity. Although the basic framework of the S-phase checkpoint in multicellular organisms has been outlined, the mechanistic details remain to be elucidated. Human chromosome-11 band-q23 translocations disrupting the MLL gene lead to poor prognostic leukaemias. Here we assign MLL as a novel effector in the mammalian S-phase checkpoint network and identify checkpoint dysfunction as an underlying mechanism of MLL leukaemias. MLL is phosphorylated at serine 516 by ATR in response to genotoxic stress in the S phase, which disrupts its interaction with, and hence its degradation by, the SCF(Skp2) E3 ligase, leading to its accumulation. Stabilized MLL protein accumulates on chromatin, methylates histone H3 lysine 4 at late replication origins and inhibits the loading of CDC45 to delay DNA replication. Cells deficient in MLL showed radioresistant DNA synthesis and chromatid-type genomic abnormalities, indicative of S-phase checkpoint dysfunction. Reconstitution of Mll(-/-) (Mll also known as Mll1) mouse embryonic fibroblasts with wild-type but not S516A or ΔSET mutant MLL rescues the S-phase checkpoint defects. Moreover, murine myeloid progenitor cells carrying an Mll-CBP knock-in allele that mimics human t(11;16) leukaemia show a severe radioresistant DNA synthesis phenotype. MLL fusions function as dominant negative mutants that abrogate the ATR-mediated phosphorylation/stabilization of wild-type MLL on damage to DNA, and thus compromise the S-phase checkpoint. Together, our results identify MLL as a key constituent of the mammalian DNA damage response pathway and show that deregulation of the S-phase checkpoint incurred by MLL translocations probably contributes to the pathogenesis of human MLL leukaemias.


Subject(s)
Cell Cycle Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase/physiology , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line , Chromatin/metabolism , DNA Damage , DNA Replication/physiology , Genes, Dominant/genetics , Genomic Instability/physiology , Histone-Lysine N-Methyltransferase , Histones/chemistry , Histones/metabolism , Humans , Leukemia/genetics , Lysine/metabolism , Methylation , Mice , Myeloid Progenitor Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Phosphorylation , Phosphoserine/metabolism , Protein Binding , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Translocation, Genetic/genetics
19.
Nature ; 458(7237): 529-33, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19212323

ABSTRACT

Epigenetic mechanisms that maintain neurogenesis throughout adult life remain poorly understood. Trithorax group (trxG) and Polycomb group (PcG) gene products are part of an evolutionarily conserved chromatin remodelling system that activate or silence gene expression, respectively. Although PcG member Bmi1 has been shown to be required for postnatal neural stem cell self-renewal, the role of trxG genes remains unknown. Here we show that the trxG member Mll1 (mixed-lineage leukaemia 1) is required for neurogenesis in the mouse postnatal brain. Mll1-deficient subventricular zone neural stem cells survive, proliferate and efficiently differentiate into glial lineages; however, neuronal differentiation is severely impaired. In Mll1-deficient cells, early proneural Mash1 (also known as Ascl1) and gliogenic Olig2 expression are preserved, but Dlx2, a key downstream regulator of subventricular zone neurogenesis, is not expressed. Overexpression of Dlx2 can rescue neurogenesis in Mll1-deficient cells. Chromatin immunoprecipitation demonstrates that Dlx2 is a direct target of MLL in subventricular zone cells. In differentiating wild-type subventricular zone cells, Mash1, Olig2 and Dlx2 loci have high levels of histone 3 trimethylated at lysine 4 (H3K4me3), consistent with their transcription. In contrast, in Mll1-deficient subventricular zone cells, chromatin at Dlx2 is bivalently marked by both H3K4me3 and histone 3 trimethylated at lysine 27 (H3K27me3), and the Dlx2 gene fails to properly activate. These data support a model in which Mll1 is required to resolve key silenced bivalent loci in postnatal neural precursors to the actively transcribed state for the induction of neurogenesis, but not for gliogenesis.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Neurogenesis , Neurons/cytology , Stem Cells/cytology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cell Survival , Cells, Cultured , Chromatin Immunoprecipitation , Histone-Lysine N-Methyltransferase , Histones/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Methylation , Mice , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Oligodendrocyte Transcription Factor 2 , Stem Cells/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Nat Rev Cancer ; 7(11): 823-33, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17957188

ABSTRACT

Translocations that involve the mixed lineage leukaemia (MLL) gene identify a unique group of acute leukaemias, and often predict a poor prognosis. The MLL gene encodes a DNA-binding protein that methylates histone H3 lysine 4 (H3K4), and positively regulates gene expression including multiple Hox genes. Leukaemogenic MLL translocations encode MLL fusion proteins that have lost H3K4 methyltransferase activity. A key feature of MLL fusion proteins is their ability to efficiently transform haematopoietic cells into leukaemia stem cells. The link between a chromatin modulator and leukaemia stem cells provides support for epigenetic landscapes as an important part of leukaemia and normal stem-cell development.


Subject(s)
Epigenesis, Genetic/physiology , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/physiology , Adult , Animals , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Child , Chromatin/ultrastructure , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Humans , Infant , Methylation , Mice , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/deficiency , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplastic Stem Cells/cytology , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Prognosis , Protein Methyltransferases , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...