Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Chembiochem ; 24(24): e202300515, 2023 12 14.
Article in English | MEDLINE | ID: mdl-37807669

ABSTRACT

NSD2 is a histone methyltransferase predominantly catalyzing di-methylation of histone H3 on lysine K36. Increased NSD2 activity due to mutations or fusion-events affecting the gene encoding NSD2 is considered an oncogenic event and a driver in various cancers, including multiple myelomas carrying t(4;14) chromosomal translocations and acute lymphoblastic leukemia's expressing the hyperactive NSD2 mutant E1099 K. Using DNA-encoded libraries, we have identified small molecule ligands that selectively and potently bind to the PWWP1 domain of NSD2, inhibit NSD2 binding to H3K36me2-bearing nucleosomes, but do not inhibit the methyltransferase activity. The ligands were subsequently converted to selective VHL1-recruiting NSD2 degraders and by using one of the most efficacious degraders in cell lines, we show that it leads to NSD2 degradation, decrease in K3 K36me2 levels and inhibition of cell proliferation.


Subject(s)
Histone-Lysine N-Methyltransferase , Histones , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Nucleosomes , Cell Line, Tumor , Methylation
2.
Hum Mutat ; 43(7): 889-899, 2022 07.
Article in English | MEDLINE | ID: mdl-35165976

ABSTRACT

Heterozygous pathogenic variants in CIC, which encodes a transcriptional repressor, have been identified in individuals with neurodevelopmental phenotypes. To date, 11 CIC variants have been associated with the CIC-related neurodevelopmental syndrome. Here, we describe three novel and one previously reported CIC variants in four individuals with neurodevelopmental delay. Notably, we report for the first time a de novo frameshift variant specific to the long isoform of CIC (CIC-L, NM_001304815.1:c.1100dup, p.Pro368AlafsTer16) in an individual with speech delay, intellectual disability, and autism spectrum disorder. Our investigation into the function of CIC-L reveals that partial loss of CIC-L leads to transcriptional derepression of CIC target genes. We also describe a missense variant (NM_015125.3:c.683G>A, p.Arg228Gln) in an individual with a history of speech delay and relapsed pre-B acute lymphoblastic leukemia. Functional studies of this variant suggest a partial loss of CIC transcriptional repressor activity. Our study expands the list of CIC pathogenic variants and contributes to the accumulating evidence that CIC haploinsufficiency or partial loss of function is a pathogenic mechanism causing neurodevelopmental phenotypes.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Language Development Disorders , Neurodevelopmental Disorders , Autism Spectrum Disorder/genetics , Heterozygote , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Language Development Disorders/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Phenotype
3.
Genome Res ; 30(8): 1119-1130, 2020 08.
Article in English | MEDLINE | ID: mdl-32747411

ABSTRACT

Polycomb group proteins are important for maintaining gene expression patterns and cell identity in metazoans. The mammalian Polycomb repressive deubiquitinase (PR-DUB) complexes catalyze removal of monoubiquitination on lysine 119 of histone H2A (H2AK119ub1) through a multiprotein core comprised of BAP1, HCFC1, FOXK1/2, and OGT in combination with either of ASXL1, 2, or 3. Mutations in PR-DUB components are frequent in cancer. However, mechanistic understanding of PR-DUB function in gene regulation is limited. Here, we show that BAP1 is dependent on the ASXL proteins and FOXK1/2 in facilitating gene activation across the genome. Although PR-DUB was previously shown to cooperate with PRC2, we observed minimal overlap and functional interaction between BAP1 and PRC2 in embryonic stem cells. Collectively, these results demonstrate that PR-DUB, by counteracting accumulation of H2AK119ub1, maintains chromatin in an optimal configuration ensuring expression of genes important for general functions such as cell metabolism and homeostasis.


Subject(s)
Chromatin/metabolism , Deubiquitinating Enzymes/metabolism , Gene Expression Regulation/genetics , Histones/metabolism , Polycomb-Group Proteins/metabolism , Animals , Cell Proliferation/genetics , Cells, Cultured , Chromatin/genetics , Deubiquitinating Enzymes/genetics , Forkhead Transcription Factors/metabolism , Gene Knockout Techniques , Mice , Mice, Inbred BALB C , Mice, Knockout , Mouse Embryonic Stem Cells , Polycomb-Group Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
4.
Cancer Res ; 80(17): 3466-3479, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32641411

ABSTRACT

The mutant protein FOXL2C134W is expressed in at least 95% of adult-type ovarian granulosa cell tumors (AGCT) and is considered to be a driver of oncogenesis in this disease. However, the molecular mechanism by which FOXL2C134W contributes to tumorigenesis is not known. Here, we show that mutant FOXL2C134W acquires the ability to bind SMAD4, forming a FOXL2C134W/SMAD4/SMAD2/3 complex that binds a novel hybrid DNA motif AGHCAHAA, unique to the FOXL2C134W mutant. This binding induced an enhancer-like chromatin state, leading to transcription of nearby genes, many of which are characteristic of epithelial-to-mesenchymal transition. FOXL2C134W also bound hybrid loci in primary AGCT. Ablation of SMAD4 or SMAD2/3 resulted in strong reduction of FOXL2C134W binding at hybrid sites and decreased expression of associated genes. Accordingly, inhibition of TGFß mitigated the transcriptional effect of FOXL2C134W. Our results provide mechanistic insight into AGCT pathogenesis, identifying FOXL2C134W and its interaction with SMAD4 as potential therapeutic targets to this condition. SIGNIFICANCE: FOXL2C134W hijacks SMAD4 and leads to the expression of genes involved in EMT, stemness, and oncogenesis in AGCT, making FOXL2C134W and the TGFß pathway therapeutic targets in this condition. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3466/F1.large.jpg.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Forkhead Box Protein L2/genetics , Gene Expression Regulation, Neoplastic/genetics , Granulosa Cell Tumor/pathology , Smad Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Female , Forkhead Box Protein L2/metabolism , Granulosa Cell Tumor/genetics , Granulosa Cell Tumor/metabolism , Humans , Mutation , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Smad4 Protein/metabolism
5.
Cancer Res ; 78(15): 4114-4125, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29844126

ABSTRACT

Oligodendrogliomas are brain tumors accounting for approximately 10% of all central nervous system cancers. CIC is a transcription factor that is mutated in most patients with oligodendrogliomas; these mutations are believed to be a key oncogenic event in such cancers. Analysis of the Drosophila melanogaster ortholog of CIC, Capicua, indicates that CIC loss phenocopies activation of the EGFR/RAS/MAPK pathway, and studies in mammalian cells have demonstrated a role for CIC in repressing the transcription of the PEA3 subfamily of ETS transcription factors. Here, we address the mechanism by which CIC represses transcription and assess the functional consequences of CIC inactivation. Genome-wide binding patterns of CIC in several cell types revealed that CIC target genes were enriched for MAPK effector genes involved in cell-cycle regulation and proliferation. CIC binding to target genes was abolished by high MAPK activity, which led to their transcriptional activation. CIC interacted with the SIN3 deacetylation complex and, based on our results, we suggest that CIC functions as a transcriptional repressor through the recruitment of histone deacetylases. Independent single amino acid substitutions found in oligodendrogliomas prevented CIC from binding its target genes. Taken together, our results show that CIC is a transcriptional repressor of genes regulated by MAPK signaling, and that ablation of CIC function leads to increased histone acetylation levels and transcription at these genes, ultimately fueling mitogen-independent tumor growth.Significance: Inactivation of CIC inhibits its direct repression of MAPK pathway genes, leading to their increased expression and mitogen-independent growth.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/15/4114/F1.large.jpg Cancer Res; 78(15); 4114-25. ©2018 AACR.


Subject(s)
Brain Neoplasms/genetics , Histone Deacetylases/genetics , MAP Kinase Signaling System/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Animals , Cell Cycle/genetics , Cell Line , Cell Proliferation/genetics , HEK293 Cells , Humans , Mice , Transcription, Genetic/genetics
6.
Mol Cell Biol ; 34(6): 1031-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24396064

ABSTRACT

The histone demethylase JMJD2C, also known as KDM4C/GASC1, has activity against methylated H3K9 and H3K36 and is amplified and/or overexpressed in human cancers. By the generation of Jmjd2c knockout mice, we demonstrate that loss of Jmjd2c is compatible with cellular proliferation, embryonic stem cell (ESC) self-renewal, and embryonic development. Moreover, we report that JMJD2C localizes to H3K4me3-positive transcription start sites in both primary cells and in the human carcinoma KYSE150 cell line containing an amplification of the JMJD2C locus. Binding is dependent on the double Tudor domain of JMJD2C, which recognizes H3K4me3 but not H4K20me2/me3 in vitro, showing a binding specificity different from that of the double Tudor domains of JMJD2A and JMJD2B. Depletion of JMJD2C in KYSE150 cells has a modest effect on H3K9me3 and H3K36me3 levels but impairs proliferation and leads to deregulated expression of a subset of target genes involved in cell cycle progression. Taking these findings together, we show that JMJD2C is targeted to H3K4me3-positive transcription start sites, where it can contribute to transcriptional regulation, and report that the putative oncogene JMJD2C generally is not required for cellular proliferation or embryonic development.


Subject(s)
Embryonic Development/genetics , Histones/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Transcription, Genetic/genetics , Animals , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Embryonic Stem Cells/metabolism , Female , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Protein Binding/genetics , Transcription Initiation Site
7.
PLoS One ; 8(7): e67653, 2013.
Article in English | MEDLINE | ID: mdl-23844048

ABSTRACT

Posttranslational modifications (PTMs) of the histone H3 tail such as methylation, acetylation and phosphorylation play important roles in epigenetic signaling. Here we study the effect of some of these PTMs on the demethylation rates of methylated lysine 9 in vitro using peptide substrates mimicking histone H3. Various combinations with other PTMs were employed to study possible cross-talk effects by comparing enzyme kinetic characteristics. We compared the kinetics of histone tail substrates for truncated histone lysine demethylases KDM4A and KDM4C containing only the catalytic core (cc) and some combinations were characterized on full length (FL) KDM4A and KDM4C. We found that the substrates combining trimethylated K4 and K9 resulted in a significant increase in the catalytic activity for FL-KDM4A. For the truncated versions of KDM4A and KDM4C a two-fold increase in the catalytic activity toward bis-trimethylated substrates could be observed. Furthermore, a significant difference in the catalytic activity between dimethylated and trimethylated substrates was found for full length demethylases in line with what has been reported previously for truncated demethylases. Histone peptide substrates phosphorylated at T11 could not be demethylated by neither truncated nor full length KDM4A and KDM4C, suggesting that phosphorylation of threonine 11 prevents demethylation of the H3K9me3 mark on the same peptide. Acetylation of K14 was also found to influence demethylation rates significantly. Thus, for truncated KDM4A, acetylation on K14 of the substrate leads to an increase in enzymatic catalytic efficiency (k cat/K m), while for truncated KDM4C it induces a decrease, primarily caused by changes in K m. This study demonstrates that demethylation activities towards trimethylated H3K9 are significantly influenced by other PTMs on the same peptide, and emphasizes the importance of studying these interactions at the peptide level to get a more detailed understanding of the dynamics of epigenetic marks.


Subject(s)
Histone Demethylases/metabolism , Histones/metabolism , Lysine/metabolism , Protein Processing, Post-Translational , Acetylation , Amino Acid Sequence , Enzyme Assays , Epigenesis, Genetic , Histone Demethylases/chemistry , Histones/chemistry , Humans , Kinetics , Lysine/chemistry , Methylation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Substrate Specificity
9.
Nature ; 473(7347): 343-8, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21490601

ABSTRACT

Enzymes catalysing the methylation of the 5-position of cytosine (mC) have essential roles in regulating gene expression and maintaining cellular identity. Recently, TET1 was found to hydroxylate the methyl group of mC, converting it to 5-hydroxymethyl cytosine (hmC). Here we show that TET1 binds throughout the genome of embryonic stem cells, with the majority of binding sites located at transcription start sites (TSSs) of CpG-rich promoters and within genes. The hmC modification is found in gene bodies and in contrast to mC is also enriched at CpG-rich TSSs. We provide evidence further that TET1 has a role in transcriptional repression. TET1 binds a significant proportion of Polycomb group target genes. Furthermore, TET1 associates and colocalizes with the SIN3A co-repressor complex. We propose that TET1 fine-tunes transcription, opposes aberrant DNA methylation at CpG-rich sequences and thereby contributes to the regulation of DNA methylation fidelity.


Subject(s)
Cytosine/analogs & derivatives , DNA Methylation , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription, Genetic , 5-Methylcytosine/analogs & derivatives , Animals , Cell Line , CpG Islands/genetics , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Down-Regulation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , Mice , Protein Binding , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transcription Initiation Site
10.
Nature ; 464(7286): 306-10, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20075857

ABSTRACT

The Polycomb group (PcG) proteins have an important role in controlling the expression of genes essential for development, differentiation and maintenance of cell fates. The Polycomb repressive complex 2 (PRC2) is believed to regulate transcriptional repression by catalysing the di- and tri-methylation of lysine 27 on histone H3 (H3K27me2/3). At present, it is unknown how the PcG proteins are recruited to their target promoters in mammalian cells. Here we show that PRC2 forms a stable complex with the Jumonji- and ARID-domain-containing protein, JARID2 (ref. 4). Using genome-wide location analysis, we show that JARID2 binds to more than 90% of previously mapped PcG target genes. Notably, we show that JARID2 is sufficient to recruit PcG proteins to a heterologous promoter, and that inhibition of JARID2 expression leads to a major loss of PcG binding and to a reduction of H3K27me3 levels on target genes. Consistent with an essential role for PcG proteins in early development, we demonstrate that JARID2 is required for the differentiation of mouse embryonic stem cells. Thus, these results demonstrate that JARID2 is essential for the binding of PcG proteins to target genes and, consistent with this, for the proper differentiation of embryonic stem cells and normal development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Gene Expression Regulation , HeLa Cells , Humans , Mice , Nerve Tissue Proteins/genetics , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Promoter Regions, Genetic , Protein Binding
11.
Genes Dev ; 23(10): 1171-6, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19451217

ABSTRACT

The tumor suppressor proteins p16INK4A and p14ARF, encoded by the INK4A-ARF locus, are key regulators of cellular senescence. The locus is epigenetically silenced by the repressive H3K27me3 mark in normally growing cells, but becomes activated in response to oncogenic stress. Here, we show that expression of the histone H3 Lys 27 (H3K27) demethylase JMJD3 is induced upon activation of the RAS-RAF signaling pathway. JMJD3 is recruited to the INK4A-ARF locus and contributes to the transcriptional activation of p16INK4A in human diploid fibroblasts. Additionally, inhibition of Jmjd3 expression in mouse embryonic fibroblasts results in suppression of p16Ink4a and p19Arf expression and in their immortalization.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation , Oncogenes/physiology , Oxidoreductases, N-Demethylating/metabolism , Stress, Physiological/physiology , Animals , Cell Line , Cell Proliferation , Fibroblasts/cytology , Fibroblasts/metabolism , Genes, p16/physiology , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/metabolism , raf Kinases/metabolism , ras Proteins/metabolism
12.
Genes Dev ; 22(10): 1345-55, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483221

ABSTRACT

Polycomb group (PcG) proteins regulate important cellular processes such as embryogenesis, cell proliferation, and stem cell self-renewal through the transcriptional repression of genes determining cell fate decisions. The Polycomb-Repressive Complex 2 (PRC2) is highly conserved during evolution, and its intrinsic histone H3 Lys 27 (K27) trimethylation (me3) activity is essential for PcG-mediated transcriptional repression. Here, we show a functional interplay between the PRC2 complex and the H3K4me3 demethylase Rbp2 (Jarid1a) in mouse embryonic stem (ES) cells. By genome-wide location analysis we found that Rbp2 is associated with a large number of PcG target genes in mouse ES cells. We show that the PRC2 complex recruits Rbp2 to its target genes, and that this interaction is required for PRC2-mediated repressive activity during ES cell differentiation. Taken together, these results demonstrate an elegant mechanism for repression of developmental genes by the coordinated regulation of epigenetic marks involved in repression and activation of transcription.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Oxidoreductases, N-Demethylating/physiology , Repressor Proteins/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins , Down-Regulation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases , Lysine/metabolism , Mice , Models, Biological , Oxidoreductases, N-Demethylating/metabolism , Polycomb-Group Proteins , Protein Binding , Protein Methyltransferases , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 2
13.
Genes Dev ; 22(9): 1115-40, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451103

ABSTRACT

The enzymes catalyzing lysine and arginine methylation of histones are essential for maintaining transcriptional programs and determining cell fate and identity. Until recently, histone methylation was regarded irreversible. However, within the last few years, several families of histone demethylases erasing methyl marks associated with gene repression or activation have been identified, underscoring the plasticity and dynamic nature of histone methylation. Recent discoveries have revealed that histone demethylases take part in large multiprotein complexes synergizing with histone deacetylases, histone methyltransferases, and nuclear receptors to control developmental and transcriptional programs. Here we review the emerging biochemical and biological functions of the histone demethylases and discuss their potential involvement in human diseases, including cancer.


Subject(s)
Histones/metabolism , Oxidoreductases, N-Demethylating/metabolism , Aging , Animals , Cell Differentiation , Humans , Methylation , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Oxidoreductases, N-Demethylating/classification , Phylogeny
14.
Curr Opin Genet Dev ; 18(2): 159-68, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18281209

ABSTRACT

Epigenetic information refers to heritable changes in gene function that are stable between cell divisions but which is not a result of changes in the DNA sequence. Part of the epigenetic mechanism has been ascribed to modifications of histones or DNA that affects the transcription of specific genes. In this context, post-translational modifications of histone tails, in particular methylation of lysines, are regarded as important for the storage of epigenetic information. Regulation of this information plays an important role during cellular differentiation where cells with different characteristic features evolve from the same ancestor, despite identical genomic material. The characterization of several enzymes catalyzing histone lysine methylation have supported this concept by showing the requirement of these enzymes for normal development and their involvement in diseases such as cancer. The recent identification of proteins with histone demethylase activity has shown that the methylated mark is much more dynamic than previously anticipated, thereby potentially challenging the concept of histone-methylation in stable epigenetic programming.


Subject(s)
Histones/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , Humans , Methylation , Oxidoreductases, N-Demethylating/classification , Oxidoreductases, N-Demethylating/genetics , Phylogeny
15.
Nature ; 449(7163): 731-4, 2007 Oct 11.
Article in English | MEDLINE | ID: mdl-17713478

ABSTRACT

The trithorax and the polycomb group proteins are chromatin modifiers, which play a key role in the epigenetic regulation of development, differentiation and maintenance of cell fates. The polycomb repressive complex 2 (PRC2) mediates transcriptional repression by catalysing the di- and tri-methylation of Lys 27 on histone H3 (H3K27me2/me3). Owing to the essential role of the PRC2 complex in repressing a large number of genes involved in somatic processes, the H3K27me3 mark is associated with the unique epigenetic state of stem cells. The rapid decrease of the H3K27me3 mark during specific stages of embryogenesis and stem-cell differentiation indicates that histone demethylases specific for H3K27me3 may exist. Here we show that the human JmjC-domain-containing proteins UTX and JMJD3 demethylate tri-methylated Lys 27 on histone H3. Furthermore, we demonstrate that ectopic expression of JMJD3 leads to a strong decrease of H3K27me3 levels and causes delocalization of polycomb proteins in vivo. Consistent with the strong decrease in H3K27me3 levels associated with HOX genes during differentiation, we show that UTX directly binds to the HOXB1 locus and is required for its activation. Finally mutation of F18E9.5, a Caenorhabditis elegans JMJD3 orthologue, or inhibition of its expression, results in abnormal gonad development. Taken together, these results suggest that H3K27me3 demethylation regulated by UTX/JMJD3 proteins is essential for proper development. Moreover, the recent demonstration that UTX associates with the H3K4me3 histone methyltransferase MLL2 (ref. 8) supports a model in which the coordinated removal of repressive marks, polycomb group displacement, and deposition of activating marks are important for the stringent regulation of transcription during cellular differentiation.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Nuclear Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , Cell Line , Gonads/cytology , Gonads/embryology , Gonads/metabolism , Histone Demethylases , Humans , Jumonji Domain-Containing Histone Demethylases , Methylation , Nuclear Proteins/genetics , Oxidoreductases, N-Demethylating/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation
16.
Cell ; 128(6): 1063-76, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17320161

ABSTRACT

Methylation of histones has been regarded as a stable modification defining the epigenetic program of the cell, which regulates chromatin structure and transcription. However, the recent discovery of histone demethylases has challenged the stable nature of histone methylation. Here we demonstrate that the JARID1 proteins RBP2, PLU1, and SMCX are histone demethylases specific for di- and trimethylated histone 3 lysine 4 (H3K4). Consistent with a role for the JARID1 Drosophila homolog Lid in regulating expression of homeotic genes during development, we show that RBP2 is displaced from Hox genes during embryonic stem (ES) cell differentiation correlating with an increase of their H3K4me3 levels and expression. Furthermore, we show that mutation or RNAi depletion of the C. elegans JARID1 homolog rbr-2 leads to increased levels of H3K4me3 during larval development and defects in vulva formation. Taken together, these results suggest that H3K4me3/me2 demethylation regulated by the JARID1 family plays an important role during development.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Carrier Proteins/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila melanogaster/enzymology , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Gene Deletion , Genes, Homeobox , Histone Demethylases , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Jumonji Domain-Containing Histone Demethylases , Lysine , Methylation , Mice , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/genetics , Phylogeny , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 2 , Schizosaccharomyces/enzymology , Sequence Alignment , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
17.
Nature ; 442(7100): 307-11, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16732293

ABSTRACT

Methylation of lysine and arginine residues on histone tails affects chromatin structure and gene transcription. Tri- and dimethylation of lysine 9 on histone H3 (H3K9me3/me2) is required for the binding of the repressive protein HP1 and is associated with heterochromatin formation and transcriptional repression in a variety of species. H3K9me3 has long been regarded as a 'permanent' epigenetic mark. In a search for proteins and complexes interacting with H3K9me3, we identified the protein GASC1 (gene amplified in squamous cell carcinoma 1), which belongs to the JMJD2 (jumonji domain containing 2) subfamily of the jumonji family, and is also known as JMJD2C. Here we show that three members of this subfamily of proteins demethylate H3K9me3/me2 in vitro through a hydroxylation reaction requiring iron and alpha-ketoglutarate as cofactors. Furthermore, we demonstrate that ectopic expression of GASC1 or other JMJD2 members markedly decreases H3K9me3/me2 levels, increases H3K9me1 levels, delocalizes HP1 and reduces heterochromatin in vivo. Previously, GASC1 was found to be amplified in several cell lines derived from oesophageal squamous carcinomas, and in agreement with a contribution of GASC1 to tumour development, inhibition of GASC1 expression decreases cell proliferation. Thus, in addition to identifying GASC1 as a histone trimethyl demethylase, we suggest a model for how this enzyme might be involved in cancer development, and propose it as a target for anti-cancer therapy.


Subject(s)
Histones/chemistry , Histones/metabolism , Lysine/chemistry , Lysine/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Cell Proliferation , HeLa Cells , Humans , Hydroxylation , Jumonji Domain-Containing Histone Demethylases , Methylation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/classification , Neoplasm Proteins/genetics , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/classification , Oncogene Proteins/genetics , Oncogenes/genetics , Protein Binding , Substrate Specificity , Transcription Factors/antagonists & inhibitors , Transcription Factors/classification , Transcription Factors/genetics
18.
Clin Lab ; 50(9-10): 585-98, 2004.
Article in English | MEDLINE | ID: mdl-15481635

ABSTRACT

INTRODUCTION: Osteocalcin (OC) is a small bone matrix protein exclusively found in mineralized tissue. OC measured in serum or plasma provides an index of bone formation. In the present study a sensitive inhibition ELISA was established that could quantify fragments derived from the OC Mid-region in human urine. METHODS: The ELISA was based on a monoclonal antibody directed against residues 21-29 of human OC (Mid-OC Urine ELISA). OC fragments were isolated from human urine by immunoaffinity chromatography. OC fragments were purified further by reversed phase high performance chromatography for characterization by N-terminal sequencing and mass-spectrometry. OC fragments were assayed in bone cell culture supernatants and in serum and urine from patients undergoing anti-resorptive bisphosphonate therapy using the Mid-OC urine ELISA. RESULTS AND CONCLUSION: It was demonstrated that the release of OC fragments was highly correlated with osteoclast-mediated pit formation (r2= 0.89) and with an established marker of bone resorption (CTX; r2=0.91). Mid-OC values were decreased markedly after 3 and 10 days of anti-resorptive bisphosphonate treatment further indicating that the marker reflects bone resorption. The molecular characterization revealed that most of these molecules were less than 15 amino acids in length and many contained modified aspartyl residues (D-aspartyl and isoaspartyl) characteristic of aged proteins. The presence of such modifications shows that these molecules have resided in the bone matrix for an extended period and thus they cannot be derived directly from bone formation. In conclusion, these findings demonstrate that OC-fragments are released during osteoclastic bone resorption and that the quantification of specific age-modified OC fragments can provide an index of bone resorption.


Subject(s)
Bone Resorption/urine , Enzyme-Linked Immunosorbent Assay/methods , Osteocalcin/urine , Peptide Fragments/urine , Adult , Bone and Bones/metabolism , Female , Humans , Male , Middle Aged , Osteitis Deformans/blood , Osteitis Deformans/urine
19.
Menopause ; 11(5): 508-18, 2004.
Article in English | MEDLINE | ID: mdl-15356403

ABSTRACT

OBJECTIVE: Several observational studies indicate that estrogen deficiency increases the incidence of osteoarthritis in postmenopausal women. To validate this observation, we investigated the effects of ovariectomy (OVX) on cartilage erosion in rats using histology and an established bio-assay of cartilage-specific collagen type II degradation products (CTX-II). Furthermore, we investigated whether estrogen and levormeloxifene, a selective estrogen-receptor modulator (SERM), can prevent the OVX-induced changes in cartilage degradation. The clinical relevance was assessed in postmenopausal women by measuring the changes in CTX-II during 12-month treatment with levormeloxifene versus placebo. DESIGN: Sixty 6-month-old rats were divided in five groups. One group was subjected to sham and the others to OVX, followed by treatment with vehicle alone, estradiol or 0.2 mg/kg/day or 5 mg/kg/day of levormeloxifene. The rats were treated for 9 weeks with biweekly blood and urine sampling for measurement of bone resorption and cartilage turnover. After study termination, hind knees were removed for histological analysis of erosions. The effect of levormeloxifene in post-menopausal women was assessed by measuring CTX-II in samples from 301 women who were participating in a phase II study of this SERM. RESULTS: OVX rats showed significant increases in the urinary excretion of CTX-II. After 9 weeks this was manifested as increased surface erosion of knee articular cartilage compared with sham-operated rats. Treatment with estrogen or levormeloxifene prevented the OVX-induced changes. There was a significant correlation between the 4-week changes in CTX-II and cartilage erosion at week 9 (r = 0.64, P < 0.001). In postmenopausal women treated with levormeloxifene, the urinary excretion of CTX-II was decreased by approximately 50% and restored CTX-II levels to the premenopausal range. CONCLUSIONS: This study is the first to demonstrate that a SERM suppresses cartilage degradation in both rodents and humans, suggesting potential therapeutical benefits in the prevention of destructive joint diseases such as osteoarthritis.


Subject(s)
Cartilage, Articular/drug effects , Estrogen Replacement Therapy/methods , Osteoporosis, Postmenopausal/prevention & control , Pyrrolidines/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Aged , Analysis of Variance , Animals , Biopsy, Needle , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Disease Models, Animal , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Middle Aged , Ovariectomy , Postmenopause , Probability , Prognosis , Rats , Rats, Sprague-Dawley , Risk Assessment , Statistics, Nonparametric , Treatment Outcome
20.
Biogerontology ; 5(3): 139-58, 2004.
Article in English | MEDLINE | ID: mdl-15190184

ABSTRACT

Proteins are complex organic molecules susceptible to numerous post-translational modifications occurring spontaneously during aging or as a consequence of physiologic or pathologic processes. Antigenicity and interactions of proteins with components of the immune system may be profoundly affected by post-translational modifications. Thus, modified self-antigens may be absent (not-tolerated) during early T-cell selection and trigger reactions by the immune system as they arise later in life. In turn, this may play a role in the initiation and pathogenesis of autoimmune diseases. This Review article presents an overview of protein modifications that have been shown to affect antigenicity and presentation of protein antigens in autoimmune diseases. The relevance of these observations is discussed, and the implications for future prophylactic and therapeutic interventions are outlined.


Subject(s)
Aging/metabolism , Antigens/immunology , Autoimmunity , Protein Processing, Post-Translational , Proteins/metabolism , Glycosylation , Humans , Isomerism , Major Histocompatibility Complex , Oxidative Stress , Phosphorylation , Proteins/immunology , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL