Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Biochim Biophys Acta ; 1849(8): 1081-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26149774

ABSTRACT

Skeletal muscle cell differentiation requires a family of proteins called myogenic regulatory factors (MRFs) to which MyoD belongs. The activity of MyoD is under epigenetic regulation, however, the molecular mechanism by which histone KMTs and KDMs regulate MyoD transcriptional activity through methylation remains to be determined. Here we provide evidence for a unique regulatory mechanism of MyoD transcriptional activity through demethylation by Jmjd2C demethylase whose level increases during muscle differentiation. G9a decreases MyoD stability via methylation-dependent MyoD ubiquitination. Jmjd2C directly associates with MyoD in vitro and in vivo to demethylate and stabilize MyoD. The hypo-methylated MyoD due to Jmjd2C is significantly more stable than hyper-methylated MyoD by G9a. Cul4/Ddb1/Dcaf1 pathway is essential for the G9a-mediated MyoD degradation in myoblasts. By the stabilization of MyoD, Jmjd2C increases myogenic conversion of mouse embryonic fibroblasts and MyoD transcriptional activity with erasing repressive H3K9me3 level at the promoter of MyoD target genes. Collectively, Jmjd2C increases MyoD transcriptional activity to facilitate skeletal muscle differentiation by increasing MyoD stability through inhibiting G9a-dependent MyoD degradation.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , MyoD Protein/metabolism , Oxidoreductases, N-Demethylating/physiology , Transcriptional Activation , Animals , Cell Differentiation/genetics , Cells, Cultured , Down-Regulation , Epigenesis, Genetic/physiology , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Muscle Development/genetics , Muscle, Skeletal/physiology , MyoD Protein/physiology , Myoblasts/physiology , Proteolysis
2.
Biochem Biophys Res Commun ; 464(2): 659-66, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26182878

ABSTRACT

Metastasis is the leading cause of death in breast cancer patients. However, until now, the mechanisms of breast cancer metastasis remain elusive. Epigenetic switch, including histone methylation or demethylation, which can either activates or represses transcription. The JARID1C is a histone demethylase that promotes cancer cell growth and is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation. But the pathogenic breadth and mechanistic aspects of this effect relative to breast cancer have not been defined. In this study, we aimed to investigate the role of JARID1C in breast cancer. In clinical breast cancer samples, we found that JARID1C expression was significantly upregulated in cancer lesions compared with paired normal breast tissues and its expression level is positively correlated with metastasis. Silencing JARID1C in breast cancer cells could inhibit cell migration and invasion. Moreover, we also found that the expression of BRMS1 was modulated by JARID1C. Silencing of JARID1C dramatically increased BRMS1 expression both at mRNA and protein level. Mechanistically, we found JARID1C exerts its function through modulation of H3K4me3 at the BRMS1 gene promoter, which was associated with inactive BRMS1 transcription. BRMS1 knockdown reversed shJARID1C-induced migration inhibition. Further, BRMS1 expression in human breast cancer is negatively correlated with JARID1C expression. Our results, for the first time, portray a pivotal role of JARID1C in regulating metastatic behaviors of breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Down-Regulation/physiology , Neoplasm Metastasis , Oxidoreductases, N-Demethylating/physiology , Repressor Proteins/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Gene Silencing , Histone Demethylases , Humans , Oxidoreductases, N-Demethylating/genetics
3.
Sci Transl Med ; 6(265): 265ra169, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25473037

ABSTRACT

Herpesviruses are highly prevalent and maintain lifelong latent reservoirs, thus posing challenges to the control of herpetic disease despite the availability of antiviral pharmaceuticals that target viral DNA replication. The initiation of herpes simplex virus infection and reactivation from latency is dependent on a transcriptional coactivator complex that contains two required histone demethylases, LSD1 (lysine-specific demethylase 1) and a member of the JMJD2 family (Jumonji C domain-containing protein 2). Inhibition of either of these enzymes results in heterochromatic suppression of the viral genome and blocks infection and reactivation in vitro. We demonstrate that viral infection can be epigenetically suppressed in three animal models of herpes simplex virus infection and disease. Treating animals with the monoamine oxidase inhibitor tranylcypromine to inhibit LSD1 suppressed viral lytic infection, subclinical shedding, and reactivation from latency in vivo. This phenotypic suppression was correlated with enhanced epigenetic suppression of the viral genome and suggests that, even during latency, the chromatin state of the virus is dynamic. Therefore, epi-pharmaceuticals may represent a promising approach to treat herpetic diseases.


Subject(s)
Epigenesis, Genetic , Herpesviridae Infections/metabolism , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/physiology , Animals , Disease Models, Animal , Female , Genome, Viral , Guinea Pigs , Histone Demethylases , Mice , Mice, Inbred BALB C , Monoamine Oxidase Inhibitors/chemistry , Phenotype , Protein Structure, Tertiary , Rabbits , Recurrence , Tranylcypromine/chemistry , Vagina/virology , Virus Activation , Virus Latency , Virus Replication/drug effects , Virus Shedding
4.
Biol Psychiatry ; 76(1): 57-65, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24183790

ABSTRACT

BACKGROUND: Memories associated with drugs of abuse, such as methamphetamine (METH), increase relapse vulnerability to substance use disorder by triggering craving. The nucleus accumbens (NAc) is essential to these drug-associated memories, but underlying mechanisms are poorly understood. Posttranslational chromatin modifications, such as histone methylation, modulate gene transcription; thus, we investigated the role of the associated epigenetic modifiers in METH-associated memory. METHODS: Conditioned place preference was used to assess the epigenetic landscape in the NAc supporting METH-associated memory (n = 79). The impact of histone methylation (H3K4me2/3) on the formation and expression of METH-associated memory was determined by focal, intra-NAc knockdown (KD) of a writer, the methyltransferase mixed-lineage leukemia 1 (Mll1) (n = 26), and an eraser, the histone lysine (K)-specific demethylase 5C (Kdm5c) (n = 38), of H3K4me2/3. RESULTS: A survey of chromatin modifications in the NAc of animals forming a METH-associated memory revealed the global induction of several modifications associated with active transcription. This correlated with a pattern of gene activation, as revealed by microarray analysis, including upregulation of oxytocin receptor (Oxtr) and FBJ osteosarcoma oncogene (Fos), the promoters of which also had increased H3K4me3. KD of Mll1 reduced H3K4me3, Fos and Oxtr levels and disrupted METH-associated memory. KD of Kdm5c resulted in hypermethylation of H3K4 and prevented the expression of METH-associated memory. CONCLUSIONS: The development and expression of METH-associated memory are supported by regulation of H3K4me2/3 levels by MLL1 and KDM5C, respectively, in the NAc. These data indicate that permissive histone methylation, and the associated epigenetic writers and erasers, represent potential targets for the treatment of substance abuse relapse, a psychiatric condition perpetuated by unwanted associative memories.


Subject(s)
Epigenesis, Genetic/drug effects , Histones/drug effects , Histones/metabolism , Memory/drug effects , Methamphetamine/pharmacology , Animals , Chromatin/metabolism , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Epigenesis, Genetic/physiology , Gene Knockdown Techniques , Histone Demethylases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/physiology , Male , Memory/physiology , Methylation/drug effects , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
5.
Nat Struct Mol Biol ; 20(12): 1346-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24304913

ABSTRACT

Mediator of DNA-damage checkpoint 1 (MDMDC1) has a central role in repair of DNA double-strand breaks (DSBs) by both homologous recombination and nonhomologous end joining, and its function is regulated by post-translational phosphorylation, ubiquitylation and sumoylation. In this issue, a new study by Watanabe et al. reveals that methylation of MDMDC1 is also critical for its function in DSB repair and specifically affects repair through BRCA1-dependent homologous recombination.


Subject(s)
BRCA1 Protein/physiology , DNA Breaks , DNA-Binding Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Nuclear Proteins/metabolism , Oxidoreductases, N-Demethylating/physiology , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Female , Humans , Ubiquitin-Protein Ligases
6.
Nat Struct Mol Biol ; 20(12): 1425-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24240613

ABSTRACT

Chromatin ubiquitylation flanking DNA double-strand breaks (DSBs), mediated by RNF8 and RNF168 ubiquitin ligases, orchestrates a two-branch pathway, recruiting repair factors 53BP1 or the RAP80-BRCA1 complex. We report that human demethylase JMJD1C regulates the RAP80-BRCA1 branch of this DNA-damage response (DDR) pathway. JMJD1C was stabilized by interaction with RNF8, was recruited to DSBs, and was required for local ubiquitylations and recruitment of RAP80-BRCA1 but not 53BP1. JMJD1C bound to RNF8 and MDC1, and demethylated MDC1 at Lys45, thereby promoting MDC1-RNF8 interaction, RNF8-dependent MDC1 ubiquitylation and recruitment of RAP80-BRCA1 to polyubiquitylated MDC1. Furthermore, JMJD1C restricted formation of RAD51 repair foci, and JMJD1C depletion caused resistance to ionizing radiation and PARP inhibitors, phenotypes relevant to aberrant loss of JMJD1C in subsets of breast carcinomas. These findings identify JMJD1C as a DDR component, with implications for genome-integrity maintenance, tumorigenesis and cancer treatment.


Subject(s)
BRCA1 Protein/physiology , DNA Breaks , DNA-Binding Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Nuclear Proteins/metabolism , Oxidoreductases, N-Demethylating/physiology , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing , BRCA1 Protein/chemistry , BRCA1 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle Proteins , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Female , HeLa Cells , Histone Chaperones , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Nuclear Proteins/chemistry , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Rad51 Recombinase/chemistry , Rad51 Recombinase/metabolism , Rad51 Recombinase/physiology , Trans-Activators/chemistry , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Ubiquitination
7.
Biochem J ; 452(1): 57-66, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23452237

ABSTRACT

The Tat (twin-arginine translocation) system is a protein targeting pathway utilized by prokaryotes and chloroplasts. Tat substrates are produced with distinctive N-terminal signal peptides and are translocated as fully folded proteins. In Escherichia coli, Tat-dependent proteins often contain redox cofactors that must be loaded before translocation. Trimethylamine N-oxide reductase (TorA) is a model bacterial Tat substrate and is a molybdenum cofactor-dependent enzyme. Co-ordination of cofactor loading and translocation of TorA is directed by the TorD protein, which is a cytoplasmic chaperone known to interact physically with the TorA signal peptide. In the present study, a pre-export TorAD complex has been characterized using biochemical and biophysical techniques, including SAXS (small-angle X-ray scattering). A stable, cofactor-free TorAD complex was isolated, which revealed a 1:1 binding stoichiometry. Surprisingly, a TorAD complex with similar architecture can be isolated in the complete absence of the 39-residue TorA signal peptide. The present study demonstrates that two high-affinity binding sites for TorD are present on TorA, and that a single TorD protein binds both of those simultaneously. Further characterization suggested that the C-terminal 'Domain IV' of TorA remained solvent-exposed in the cofactor-free pre-export TorAD complex. It is possible that correct folding of Domain IV upon cofactor loading is the trigger for TorD release and subsequent export of TorA.


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry , Molecular Chaperones/chemistry , Oxidoreductases, N-Demethylating/chemistry , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Membrane Transport Proteins/genetics , Molecular Chaperones/genetics , Molecular Chaperones/physiology , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/physiology , Protein Binding/genetics , Protein Sorting Signals/genetics , Protein Transport/genetics , Translocation, Genetic
8.
Development ; 140(5): 1014-23, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364332

ABSTRACT

Adult stem cells reside in microenvironments called niches, where they are regulated by both extrinsic cues, such as signaling from neighboring cells, and intrinsic factors, such as chromatin structure. Here we report that in the Drosophila testis niche an H3K27me3-specific histone demethylase encoded by Ubiquitously transcribed tetratricopeptide repeat gene on the X chromosome (dUTX) maintains active transcription of the Suppressor of cytokine signaling at 36E (Socs36E) gene by removing the repressive H3K27me3 modification near its transcription start site. Socs36E encodes an inhibitor of the Janus kinase signal transducer and activator of transcription (JAK-STAT) signaling pathway. Whereas much is known about niche-to-stem cell signaling, such as the JAK-STAT signaling that is crucial for stem cell identity and activity, comparatively little is known about signaling from stem cells to the niche. Our results reveal that stem cells send feedback to niche cells to maintain the proper gene expression and architecture of the niche. We found that dUTX acts in cyst stem cells to maintain gene expression in hub cells through activating Socs36E transcription and preventing hyperactivation of JAK-STAT signaling. dUTX also acts in germline stem cells to maintain hub structure through regulating DE-Cadherin levels. Therefore, our findings provide new insights into how an epigenetic factor regulates crosstalk among different cell types within an endogenous stem cell niche, and shed light on the biological functions of a histone demethylase in vivo.


Subject(s)
Drosophila Proteins/physiology , Gene Expression Regulation, Developmental , Janus Kinases/antagonists & inhibitors , Oxidoreductases, N-Demethylating/physiology , STAT Transcription Factors/antagonists & inhibitors , Stem Cell Niche/genetics , Testis/cytology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Adult Stem Cells/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Germ Cells/physiology , Histone Demethylases/physiology , Janus Kinases/metabolism , Male , Models, Biological , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Stem Cell Niche/physiology , Testis/metabolism , Testis/physiology
9.
Biochem Pharmacol ; 85(8): 1182-94, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23395694

ABSTRACT

The common marmoset is a promising species for evaluating the safety of drug candidates. To further understand the capacity for drug metabolism in marmosets, a cDNA encoding a CYP2B enzyme was cloned from the total RNA fraction of marmoset liver by 3'- and 5'-RACE methods. Nucleotide and deduced amino acid sequences showed 90.8 and 86.2% identity, respectively, with human CYP2B6. The marmoset CYP2B6 (marCYP2B6) protein was expressed in insect cells, and its enzymatic properties were compared with those of human (humCYP2B6) and cynomolgus monkey (cynCYP2B6) orthologs in liver and insect cell microsomes. Enzymatic functions were examined for the oxidation of 7-ethoxy-4-(trifluoromethyl)coumarin (7-ETC), bupropion (BUP) and efavirenz (EFV). The kinetic profiles for the oxidation of the three substrates by liver microsomal fractions were similar between humans and cynomolgus monkeys (biphasic for 7-ETC and monophasic for BUP and EFV), but that of marmosets was unique (monophasic for 7-ETC and biphasic for BUP and EFV). Recombinant enzymes, humCYP2B6 and cynCYP2B6, also yielded similar kinetic profiles for the oxidation of the three substrates, whereas marCYP2B6 showed activity only for 7-ETC hydroxylation. In silico docking simulations suggested that two amino acid residues, Val-114 and Leu-367, affect the activity of marCYP2B6. In fact, a marCYP2B6 mutant with substitutions V114I and L367V exhibited BUP hydroxylase activity that was 4-fold higher than that of humCYP2B6, while its EFV 8-hydroxylase activity was only 10% that of the human enzyme. These results indicate that the amino acids at positions 114 and 367 affect the enzymatic capacity of marmoset CYP2B6.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/physiology , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/physiology , Alkynes , Amino Acid Sequence , Animals , Aryl Hydrocarbon Hydroxylases/chemistry , Base Sequence , Benzoxazines/metabolism , Bupropion/metabolism , Callithrix , Cloning, Molecular , Cyclopropanes , Cytochrome P-450 CYP2B6 , Humans , Hydroxylation , Insecta , Macaca fascicularis , Microsomes, Liver/metabolism , Molecular Sequence Data , Oxidoreductases, N-Demethylating/chemistry , Structure-Activity Relationship
10.
Drug Metabol Drug Interact ; 27(4): 185-97, 2012.
Article in English | MEDLINE | ID: mdl-23152403

ABSTRACT

Cytochrome P450 (CYP) 2B6 belongs to the set of important hepatic drug-metabolizing CYPs. It makes up roughly 3%-6% of total hepatic CYP content and metabolizes several pharmaceuticals including bupropion, efavirenz, cyclophosphamide, pethidine, ketamine and propofol. The enzyme is susceptible to drug-drug interactions by enzyme induction and inhibition. In addition to drugs, CYP2B6 is able to both detoxify and bioactivate a number of procarcinogens and environmental agents including pesticides and herbicides. There is an extensive interindividual variability in the expression of CYP2B6, which is in part explained by extensive genetic polymorphism. CYP2B6 is one of the most polymorphic CYP genes in humans with over 100 described SNPs, numerous complex haplotypes and distinct ethnic and racial frequencies. This review summarizes the basic properties of CYP2B6 and the main characteristics of clinical relevance.


Subject(s)
Aryl Hydrocarbon Hydroxylases/physiology , Oxidoreductases, N-Demethylating/physiology , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP2B6 , Drug Interactions , Humans , Oxidoreductases, N-Demethylating/genetics , Pharmacogenetics
11.
Cell Mol Life Sci ; 69(24): 4177-90, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22802127

ABSTRACT

Estrogen (E(2)) regulates spermatogenesis, yet its direct target genes have not been identified in the testis. Here, we cloned the proximal 5' flanking region of the mouse fatty acid amide hydrolase (faah) gene upstream of the luciferase reporter gene, and demonstrated its promoter activity and E(2) inducibility in primary mouse Sertoli cells. Specific mutations in the E(2) response elements (ERE) of the faah gene showed that two proximal ERE sequences (ERE2/3) are essential for E(2)-induced transcription, and chromatin immunoprecipitation experiments showed that E(2) induced estrogen receptor ß binding at ERE2/3 sites in the faah promoter in vivo. Moreover, the histone demethylase LSD1 was found to be associated with ERE2/3 sites and to play a role in mediating E(2) induction of FAAH expression. E(2) induced epigenetic modifications at the faah proximal promoter compatible with transcriptional activation by remarkably decreasing methylation of both DNA at CpG site and histone H3 at lysine 9. Finally, FAAH silencing abolished E(2) protection against apoptosis induced by the FAAH substrate anandamide. Taken together, our results identify FAAH as the first direct target of E(2).


Subject(s)
Amidohydrolases/genetics , Estrogens/pharmacology , Gene Expression Regulation , Oxidoreductases, N-Demethylating/physiology , Sertoli Cells/metabolism , Amidohydrolases/chemistry , Amidohydrolases/physiology , Animals , Apoptosis , Base Sequence , DNA Methylation/drug effects , Estrogen Receptor beta/metabolism , Estrogen Receptor beta/physiology , Histone Demethylases , Histones/metabolism , Male , Methylation , Mice , Molecular Sequence Data , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic , Sertoli Cells/drug effects
12.
Leukemia ; 26(9): 2039-51, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22699452

ABSTRACT

Lysine (K)-specific demethylase 1A (LSD1/KDM1A) has been identified as a potential therapeutic target in solid cancers and more recently in acute myeloid leukemia. However, the potential side effects of a LSD1-inhibitory therapy remain elusive. Here, we show, with a newly established conditional in vivo knockdown model, that LSD1 represents a central regulator of hematopoietic stem and progenitor cells. LSD1 knockdown (LSD1-kd) expanded progenitor numbers by enhancing their proliferative behavior. LSD1-kd led to an extensive expansion of granulomonocytic, erythroid and megakaryocytic progenitors. In contrast, terminal granulopoiesis, erythropoiesis and platelet production were severely inhibited. The only exception was monopoiesis, which was promoted by LSD1 deficiency. Importantly, we showed that peripheral blood granulocytopenia, monocytosis, anemia and thrombocytopenia were reversible after LSD1-kd termination. Extramedullary splenic hematopoiesis contributed to the phenotypic reversion, and progenitor populations remained expanded. LSD1-kd was associated with the upregulation of key hematopoietic genes, including Gfi1b, Hoxa9 and Meis1, which are known regulators of the HSC/progenitor compartment. We also demonstrated that LSD1-kd abrogated Gfi1b-negative autoregulation by crossing LSD1-kd with Gfi1b:GFP mice. Taken together, our findings distinguish LSD1 as a critical regulator of hematopoiesis and point to severe, but reversible, side effects of a LSD1-targeted therapy.


Subject(s)
Cell Differentiation , Cell Proliferation , Hematopoiesis/physiology , Oxidoreductases, N-Demethylating/physiology , Stem Cells/cytology , Animals , Blotting, Western , Erythropoiesis/physiology , Female , Flow Cytometry , Granulocytes/cytology , Granulocytes/metabolism , Histone Demethylases , Humans , Integrases/metabolism , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Transgenic , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism
13.
Cancer Cell ; 21(4): 473-87, 2012 Apr 17.
Article in English | MEDLINE | ID: mdl-22464800

ABSTRACT

Using a mouse model of human MLL-AF9 leukemia, we identified the lysine-specific demethylase KDM1A (LSD1 or AOF2) as an essential regulator of leukemia stem cell (LSC) potential. KDM1A acts at genomic loci bound by MLL-AF9 to sustain expression of the associated oncogenic program, thus preventing differentiation and apoptosis. In vitro and in vivo pharmacologic targeting of KDM1A using tranylcypromine analogs active in the nanomolar range phenocopied Kdm1a knockdown in both murine and primary human AML cells exhibiting MLL translocations. By contrast, the clonogenic and repopulating potential of normal hematopoietic stem and progenitor cells was spared. Our data establish KDM1A as a key effector of the differentiation block in MLL leukemia, which may be selectively targeted to therapeutic effect.


Subject(s)
Gene Expression Regulation, Neoplastic , Histone Demethylases/physiology , Leukemia/genetics , Neoplastic Stem Cells/enzymology , Oxidoreductases, N-Demethylating/physiology , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Epigenesis, Genetic , Gene Knockdown Techniques , Histone Demethylases/genetics , Humans , Leukemia/enzymology , Leukemia/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/genetics , Oxidoreductases, N-Demethylating/genetics
14.
Cereb Cortex ; 22(6): 1431-41, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21878487

ABSTRACT

The development of a neuron from a precursor cell comprises a complex set of steps ranging from regulation of the proliferative cycle through the acquisition of distinct morphology and functionality. How these processes are orchestrated is largely unknown. Using in utero manipulation of gene expression in the mouse embryonic cerebral cortex, we found that the transition between multipolar and bipolar stages of newborn cortical pyramidal neurons is markedly delayed by depletion of CoREST, a corepressor component of chromatin remodeling complexes. This profoundly affects the onset of their radial migration. The loss of CoREST function also perturbs the dynamics of neuronal precursor cell populations, transiently increasing the fraction of cells remaining in progenitor states, but not the acquisition of the neuronal glutamatergic fate of pyramidal cells. The function of CoREST in these processes appears to be independent of its best-known interactor, the RE-1 silencer of transcription/neural restrictive silencing factor, and requires the histone demethylase LSD1. This reveals the importance of epigenetic control in the execution of neural development programs, specifically in the cerebral cortex.


Subject(s)
Cerebral Cortex/embryology , Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Oxidoreductases, N-Demethylating/physiology , Pyramidal Cells/embryology , Repressor Proteins/physiology , Animals , Cell Movement/physiology , Cerebral Cortex/cytology , Co-Repressor Proteins , Epigenesis, Genetic/physiology , Female , Histone Demethylases , Membrane Proteins/physiology , Mice , Neurons/physiology , Pregnancy
15.
Nat Struct Mol Biol ; 18(8): 867-74, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725293

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is an extreme example of cell plasticity that is important for normal development, injury repair and malignant progression. Widespread epigenetic reprogramming occurs during stem cell differentiation and malignant transformation, but EMT-related epigenetic reprogramming is poorly understood. Here we investigated epigenetic modifications during EMT mediated by transforming growth factor beta. Although DNA methylation was unchanged during EMT, we found a global reduction in the heterochromatin mark H3 Lys9 dimethylation (H3K9Me2), an increase in the euchromatin mark H3 Lys4 trimethylation (H3K4Me3) and an increase in the transcriptional mark H3 Lys36 trimethylation (H3K36Me3). These changes depended largely on lysine-specific demethylase-1 (Lsd1), and loss of Lsd1 function had marked effects on EMT-driven cell migration and chemoresistance. Genome-scale mapping showed that chromatin changes were mainly specific to large organized heterochromatin K9 modifications (LOCKs), which suggests that EMT is characterized by reprogramming of specific chromatin domains across the genome.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Animals , Cell Line , Cell Movement/genetics , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Histone Demethylases , Histones/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/physiology , Transforming Growth Factor beta/pharmacology
16.
Mol Cell ; 42(5): 689-99, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21596603

ABSTRACT

Epigenetic regulation of gene expression by histone-modifying corepressor complexes is central to normal animal development. The NAD(+)-dependent deacetylase and gene repressor SIRT1 removes histone H4K16 acetylation marks and facilitates heterochromatin formation. However, the mechanistic contribution of SIRT1 to epigenetic regulation at euchromatic loci and whether it acts in concert with other chromatin-modifying activities to control developmental gene expression programs remain unclear. We describe here a SIRT1 corepressor complex containing the histone H3K4 demethylase LSD1/KDM1A and several other LSD1-associated proteins. SIRT1 and LSD1 interact directly and play conserved and concerted roles in H4K16 deacetylation and H3K4 demethylation to repress genes regulated by the Notch signaling pathway. Mutations in Drosophila SIRT1 and LSD1 orthologs result in similar developmental phenotypes and genetically interact with the Notch pathway in Drosophila. These findings offer new insights into conserved mechanisms of epigenetic gene repression and regulation of development by SIRT1 in metazoans.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Oxidoreductases, N-Demethylating/physiology , Receptors, Notch/genetics , Sirtuin 1/physiology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histones/metabolism , Immunoprecipitation , Mutation , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Phenotype , Receptors, Notch/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
17.
Dev Biol ; 350(2): 534-47, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21146519

ABSTRACT

The conserved SWI/SNF chromatin remodeling complex uses the energy from ATP hydrolysis to alter local chromatin environments through disrupting DNA-histone contacts. These alterations influence transcription activation, as well as repression. The Drosophila SWI/SNF counterpart, known as the Brahma or Brm complex, has been shown to have an essential role in regulating the proper expression of many developmentally important genes, including those required for eye and wing tissue morphogenesis. A temperature sensitive mutation in one of the core complex subunits, SNR1 (SNF5/INI1/SMARCB1), results in reproducible wing patterning phenotypes that can be dominantly enhanced and suppressed by extragenic mutations. SNR1 functions as a regulatory subunit to modulate chromatin remodeling activities of the Brahma complex on target genes, including both activation and repression. To help identify gene targets and cofactors of the Brahma complex, we took advantage of the weak dominant nature of the snr1(E1) mutation to carry out an unbiased genetic modifier screen. Using a set of overlapping chromosomal deficiencies that removed the majority of the Drosophila genome, we looked for genes that when heterozygous would function to either enhance or suppress the snr1(E1) wing pattern phenotype. Among potential targets of the Brahma complex, we identified components of the Notch, EGFR and DPP signaling pathways important for wing development. Mutations in genes encoding histone demethylase enzymes were identified as cofactors of Brahma complex function. In addition, we found that the Lysine Specific Demethylase 1 gene (lsd1) was important for the proper cell type-specific development of wing patterning.


Subject(s)
Co-Repressor Proteins/physiology , Drosophila Proteins/physiology , Drosophila/growth & development , Oxidoreductases, N-Demethylating/physiology , Ribonucleoprotein, U1 Small Nuclear/physiology , Wings, Animal/growth & development , Animals , Cell Cycle Proteins/physiology , Cells, Cultured , Transcription Factors/physiology
18.
FEBS J ; 276(16): 4304-12, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19624733

ABSTRACT

Lysine-specific demethylase 1 (LSD1) is an enzyme that removes methyl groups from mono- and dimethylated Lys4 of histone H3, a post-translational modification associated with gene activation. Human LSD1 was the first histone demethylase to be discovered and this enzymatic activity is conserved among eukaryotes. LSD1 has been identified in a number of chromatin-remodeling complexes that control gene transcription and its demethylase activity has also been linked to pathological processes including tumorigenesis. The 852-residue sequence of LSD1 comprises an amine oxidase domain which identifies a family of enzymes that catalyze the FAD-dependent oxidation of amine substrates ranging from amino acids to aromatic neurotransmitters. Among these proteins, LSD1 is peculiar in that it acts on a protein substrate in the nuclear environment of chromatin-remodeling complexes. This functional divergence occurred during evolution from the eubacteria to eukaryotes by acquisition of additional domains such as the SWIRM domain. The N-terminal part of LSD1, predicted to be disordered, contains linear motifs that might represent functional sites responsible for the association of this enzyme with a variety of transcriptional protein complexes. LSD1 shares structural features with other flavin amine oxidases, including the overall fold of the amine oxidase domain region and details in the active site that are relevant for amine substrate oxidation.


Subject(s)
Chromatin/metabolism , Flavoproteins/physiology , Gene Expression Regulation , Oxidoreductases, N-Demethylating/physiology , Animals , Bacterial Proteins , Fungal Proteins , Histone Demethylases , Humans , Methylation , Oxidoreductases Acting on CH-NH2 Group Donors , Plant Proteins
20.
Proc Natl Acad Sci U S A ; 106(8): 2641-6, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19202064

ABSTRACT

The histone H3 demethylase Not dead yet-1 (Ndy1/KDM2B) is a physiological inhibitor of senescence. Here, we show that Ndy1 is down-regulated during senescence in mouse embryonic fibroblasts (MEFs) and that it represses the Ink4a/Arf locus. Ndy1 counteracts the senescence-associated down-regulation of Ezh2, a component of polycomb-repressive complex (PRC) 2, via a JmjC domain-dependent process leading to the global and Ink4a/Arf locus-specific up-regulation of histone H3K27 trimethylation. The latter promotes the Ink4a/Arf locus-specific binding of Bmi1, a component of PRC1. Ndy1, which interacts with Ezh2, also binds the Ink4a/Arf locus and demethylates the locus-associated histone H3K36me2 and histone H3K4me3. The combination of histone modifications driven by Ndy1 interferes with the binding of RNA Polymerase II, resulting in the transcriptional silencing of the Ink4a/Arf locus and contributing to the Ndy1 immortalization phenotype. Other studies show that, in addition to inhibiting replicative senescence, Ndy1 inhibits Ras oncogene-induced senescence via a similar molecular mechanism.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Developmental/physiology , Oxidoreductases, N-Demethylating/physiology , Animals , Cell Line, Transformed , Cellular Senescence , Enhancer of Zeste Homolog 2 Protein , Fibroblasts/cytology , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Methylation , Mice , Nuclear Proteins/physiology , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...