Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 293(50): 19250-19262, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30337366

ABSTRACT

Different transcription factors operate together at promoters and enhancers to regulate gene expression. Transcription factors either bind directly to their target DNA or are tethered to it by other proteins. The transcription factor Sp2 serves as a paradigm for indirect genomic binding. It does not require its DNA-binding domain for genomic DNA binding and occupies target promoters independently of whether they contain a cognate DNA-binding motif. Hence, Sp2 is strikingly different from its closely related paralogs Sp1 and Sp3, but how Sp2 recognizes its targets is unknown. Here, we sought to gain more detailed insights into the genomic targeting mechanism of Sp2. ChIP-exo sequencing in mouse embryonic fibroblasts revealed genomic binding of Sp2 to a composite motif where a recognition sequence for TALE homeoproteins and a recognition sequence for the trimeric histone-fold domain protein nuclear transcription factor Y (Nf-y) are separated by 11 bp. We identified a complex consisting of the TALE homeobox protein Prep1, its partner PBX homeobox 1 (Pbx1), and Nf-y as the major partners in Sp2-promoter interactions. We found that the Pbx1:Prep1 complex together with Nf-y recruits Sp2 to co-occupied regulatory elements. In turn, Sp2 potentiates binding of Pbx1:Prep1 and Nf-y. We also found that the Sp-box, a short sequence motif close to the Sp2 N terminus, is crucial for Sp2's cofactor function. Our findings reveal a mechanism by which the DNA binding-independent activity of Sp2 potentiates genomic loading of Pbx1:Prep1 and Nf-y to composite motifs present in many promoters of highly expressed genes.


Subject(s)
CCAAT-Binding Factor/metabolism , Genomics , Homeodomain Proteins/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Sp2 Transcription Factor/metabolism , Animals , CCAAT-Binding Factor/chemistry , Cell Line , Histones/metabolism , Mice , Nucleotide Motifs , Protein Binding , Protein Transport , Sp2 Transcription Factor/chemistry , Zinc Fingers
2.
PLoS Genet ; 14(1): e1007193, 2018 01.
Article in English | MEDLINE | ID: mdl-29381691

ABSTRACT

Diverse Polycomb repressive complexes 1 (PRC1) play essential roles in gene regulation, differentiation and development. Six major groups of PRC1 complexes that differ in their subunit composition have been identified in mammals. How the different PRC1 complexes are recruited to specific genomic sites is poorly understood. The Polycomb Ring finger protein PCGF6, the transcription factors MGA and E2F6, and the histone-binding protein L3MBTL2 are specific components of the non-canonical PRC1.6 complex. In this study, we have investigated their role in genomic targeting of PRC1.6. ChIP-seq analysis revealed colocalization of MGA, L3MBTL2, E2F6 and PCGF6 genome-wide. Ablation of MGA in a human cell line by CRISPR/Cas resulted in complete loss of PRC1.6 binding. Rescue experiments revealed that MGA recruits PRC1.6 to specific loci both by DNA binding-dependent and by DNA binding-independent mechanisms. Depletion of L3MBTL2 and E2F6 but not of PCGF6 resulted in differential, locus-specific loss of PRC1.6 binding illustrating that different subunits mediate PRC1.6 loading to distinct sets of promoters. Mga, L3mbtl2 and Pcgf6 colocalize also in mouse embryonic stem cells, where PRC1.6 has been linked to repression of germ cell-related genes. Our findings unveil strikingly different genomic recruitment mechanisms of the non-canonical PRC1.6 complex, which specify its cell type- and context-specific regulatory functions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , DNA/metabolism , E2F6 Transcription Factor/physiology , Nuclear Proteins/physiology , Polycomb Repressive Complex 1/metabolism , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , E2F6 Transcription Factor/genetics , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Human Embryonic Stem Cells/physiology , Humans , Mice , Mouse Embryonic Stem Cells/physiology , Nuclear Proteins/genetics , Protein Binding/genetics , Transcription Factors/genetics
3.
Biochim Biophys Acta Gene Regul Mech ; 1860(5): 590-597, 2017 May.
Article in English | MEDLINE | ID: mdl-27697431

ABSTRACT

For long it has been recognized that CCAAT boxes and GC-rich elements co-occur in many human and murine promoters within 100bp upstream of the transcription start site. The trimeric transcription factor NF-Y is the major CCAAT box-binding factor, and members of the SP family of transcription factors are the major GC box-binding proteins. Recent chromatin immunoprecipitations coupled with high throughput sequencing (ChIP-seq) have examined binding of NF-Y and the ubiquitous SP factors SP1, SP2 and SP3 genome-wide, allowing for comprehensive comparison of NF-Y and SP factor actions in the context of chromatin. Here, I attempt a synthesis of the earlier single-promoter type of analysis with the more recent genome-wide studies. In particular, I also discuss different modes of genomic interactions between SP factors and NF-Y that have emerged recently, and identify a key technical issue, which needs to be taken into account in a critical evaluation of genome-wide studies. This article is part of a Special Issue entitled: Nuclear Factor Y in Development and Disease, edited by Prof. Roberto Mantovani.


Subject(s)
CCAAT-Binding Factor/metabolism , Response Elements/physiology , Sp Transcription Factors/metabolism , Animals , CCAAT-Binding Factor/genetics , Chromatin Immunoprecipitation , Genome-Wide Association Study , Humans , Mice , Sp Transcription Factors/genetics
4.
J Neurochem ; 140(2): 245-256, 2017 01.
Article in English | MEDLINE | ID: mdl-27889927

ABSTRACT

Oligodendrocytes and Schwann cells are the myelinating glia of the vertebrate nervous system and by generation of myelin sheaths allow rapid saltatory conduction. Previous in vitro work had pointed to a role of the zinc finger containing specificity proteins Sp1 and Sp3 as major regulators of glial differentiation and myelination. Here, we asked whether such a role is also evident in vivo using mice with specific deletions of Sp1 or Sp3 in myelinating glia. We also studied glia-specific conditional Sp2- and constitutive Sp4-deficient mice to include all related glutamine-rich Sp factors into our analysis. Surprisingly, we did not detect developmental Schwann cell abnormalities in any of the mutant mice. Oligodendrocyte development and differentiation was also not fundamentally affected as oligodendrocytes were present in all mouse mutants and retained their ability to differentiate and initiate myelin gene expression. The most severe defect we observed was a 50% reduction in Mbp- and proteolipid protein 1 (Plp1)-positive differentiating oligodendrocytes in Sp2 mutants at birth. Unexpectedly, glial development appeared undisturbed even in the joint absence of Sp1 and Sp3. We conclude that Sp2 has a minor effect on the differentiation of myelinating glia, and that glutamine-rich Sp proteins are not essential regulators of the process.


Subject(s)
Cell Differentiation/physiology , Glutamine/metabolism , Myelin Sheath/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Sp2 Transcription Factor/metabolism , Animals , Cells, Cultured , Gene Expression Regulation, Developmental/physiology , Myelin Basic Protein/metabolism , Rats , Schwann Cells/drug effects , Schwann Cells/metabolism
5.
PLoS Genet ; 11(3): e1005102, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25793500

ABSTRACT

Transcription factors are grouped into families based on sequence similarity within functional domains, particularly DNA-binding domains. The Specificity proteins Sp1, Sp2 and Sp3 are paradigmatic of closely related transcription factors. They share amino-terminal glutamine-rich regions and a conserved carboxy-terminal zinc finger domain that can bind to GC rich motifs in vitro. All three Sp proteins are ubiquitously expressed; yet they carry out unique functions in vivo raising the question of how specificity is achieved. Crucially, it is unknown whether they bind to distinct genomic sites and, if so, how binding site selection is accomplished. In this study, we have examined the genomic binding patterns of Sp1, Sp2 and Sp3 in mouse embryonic fibroblasts by ChIP-seq. Sp1 and Sp3 essentially occupy the same promoters and localize to GC boxes. The genomic binding pattern of Sp2 is different; Sp2 primarily localizes at CCAAT motifs. Consistently, re-expression of Sp2 and Sp3 mutants in corresponding knockout MEFs revealed strikingly different modes of genomic binding site selection. Most significantly, while the zinc fingers dictate genomic binding of Sp3, they are completely dispensable for binding of Sp2. Instead, the glutamine-rich amino-terminal region is sufficient for recruitment of Sp2 to its target promoters in vivo. We have identified the trimeric histone-fold CCAAT box binding transcription factor Nf-y as the major partner for Sp2-chromatin interaction. Nf-y is critical for recruitment of Sp2 to co-occupied regulatory elements. Equally, Sp2 potentiates binding of Nf-y to shared sites indicating the existence of an extensive Sp2-Nf-y interaction network. Our results unveil strikingly different recruitment mechanisms of Sp1/Sp2/Sp3 transcription factor members uncovering an unexpected layer of complexity in their binding to chromatin in vivo.


Subject(s)
Protein Interaction Maps/genetics , Sp1 Transcription Factor/genetics , Sp2 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Zinc Fingers/genetics , Animals , Binding Sites , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genome , Histones/genetics , Mice , Nucleotide Motifs/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid/genetics , Sp1 Transcription Factor/metabolism , Sp2 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism
6.
Blood ; 125(12): 1957-67, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25538045

ABSTRACT

Sp1 and Sp3 belong to the specificity proteins (Sp)/Krüppel-like transcription factor family. They are closely related, ubiquitously expressed, and recognize G-rich DNA motifs. They are thought to regulate generic processes such as cell-cycle and growth control, metabolic pathways, and apoptosis. Ablation of Sp1 or Sp3 in mice is lethal, and combined haploinsufficiency results in hematopoietic defects during the fetal stages. Here, we show that in adult mice, conditional pan-hematopoietic (Mx1-Cre) ablation of either Sp1 or Sp3 has minimal impact on hematopoiesis, whereas the simultaneous loss of Sp1 and Sp3 results in severe macrothrombocytopenia. This occurs in a cell-autonomous manner as shown by megakaryocyte-specific (Pf4-Cre) double-knockout mice. We employed flow cytometry, cell culture, and electron microscopy and show that although megakaryocyte numbers are normal in bone marrow and spleen, they display a less compact demarcation membrane system and a striking inability to form proplatelets. Through megakaryocyte transcriptomics and platelet proteomics, we identified several cytoskeleton-related proteins and downstream effector kinases, including Mylk, that were downregulated upon Sp1/Sp3 depletion, providing an explanation for the observed defects in megakaryopoiesis. Supporting this notion, selective Mylk inhibition by ML7 affected proplatelet formation and stabilization and resulted in defective ITAM receptor-mediated platelet aggregation.


Subject(s)
Blood Platelets/cytology , Megakaryocytes/cytology , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Animals , Azepines/chemistry , Blood Platelets/metabolism , Bone Marrow/metabolism , Flow Cytometry , Lectins, C-Type/metabolism , Mice , Mice, Knockout , Naphthalenes/chemistry , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Proteome , Signal Transduction , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Spleen/metabolism , Thrombocytopenia/metabolism , Transcription Factors/metabolism
7.
Nucleic Acids Res ; 42(5): 3044-58, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24369422

ABSTRACT

Lethal(3) malignant brain tumour like 2 (L3MBTL2) is an integral component of the polycomb repressive complex 1.6 (PRC1.6) and has been implicated in transcriptional repression and chromatin compaction. Here, we show that L3MBTL2 is modified by SUMO2/3 at lysine residues 675 and 700 close to the C-terminus. SUMOylation of L3MBTL2 neither affected its repressive activity in reporter gene assays nor it's binding to histone tails in vitro. In order to analyse whether SUMOylation affects binding of L3MBTL2 to chromatin, we performed ChIP-Seq analysis with chromatin of wild-type HEK293 cells and with chromatin of HEK293 cells stably expressing either FLAG-tagged SUMOylation-competent or SUMOylation-defective L3MBTL2. Wild-type FLAG-L3MBTL2 and the SUMOylation-defective FLAG-L3MBTL2 K675/700R mutant essentially occupied the same sites as endogenous L3MBTL2 suggesting that SUMOylation of L3MBTL2 does not affect chromatin binding. However, a subset of L3MBTL2-target genes, particularly those with low L3MBTL2 occupancy including pro-inflammatory genes, was de-repressed in cells expressing the FLAG-L3MBTL2 K675/700R mutant. Finally, we provide evidence that SUMOylation of L3MBTL2 facilitates repression of these PRC1.6-target genes by balancing the local H2Aub1 levels established by the ubiquitinating enzyme RING2 and the de-ubiquitinating PR-DUB complex.


Subject(s)
Gene Expression Regulation , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Sumoylation , Transcription Factors/metabolism , Transcription, Genetic , Binding Sites , E2F6 Transcription Factor/metabolism , Genome, Human , HEK293 Cells , Histones/metabolism , Humans , Lysine/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Ubiquitin-Protein Ligases/metabolism
8.
PLoS One ; 8(6): e66947, 2013.
Article in English | MEDLINE | ID: mdl-23826177

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear receptor regulating adipogenesis, glucose homeostasis and inflammatory responses. The activity of PPARγ is controlled by post-translational modifications including SUMOylation and phosphorylation that affects its biological and molecular functions. Several important aspects of PPARγ SUMOylation including SUMO isoform-specificity and the impact of ligand binding on SUMOylation remain unresolved or contradictory. Here, we present a comprehensive study of PPARγ1 SUMOylation. We show that PPARγ1 can be modified by SUMO1 and SUMO2. Mutational analyses revealed that SUMOylation occurs exclusively within the N-terminal activation function 1 (AF1) domain predominantly at lysines 33 and 77. Ligand binding to the C-terminal ligand-binding domain (LBD) of PPARγ1 reduces SUMOylation of lysine 33 but not of lysine 77. SUMOylation of lysine 33 and lysine 77 represses basal and ligand-induced activation by PPARγ1. We further show that lysine 365 within the LBD is not a target for SUMOylation as suggested in a previous report, but it is essential for full LBD activity. Our results suggest that PPARγ ligands negatively affect SUMOylation by interdomain communication between the C-terminal LBD and the N-terminal AF1 domain. The ability of the LBD to regulate the AF1 domain may have important implications for the evaluation and mechanism of action of therapeutic ligands that bind PPARγ.


Subject(s)
PPAR gamma/chemistry , PPAR gamma/metabolism , Sumoylation , HEK293 Cells , HeLa Cells , Humans , Ligands , Lysine/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/metabolism , Serine/genetics , Small Ubiquitin-Related Modifier Proteins/metabolism , Structure-Activity Relationship , Transcription, Genetic
9.
Nucleic Acids Res ; 40(16): 7844-57, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684502

ABSTRACT

The transcription factor Sp2 is essential for early mouse development and for proliferation of mouse embryonic fibroblasts in culture. Yet its mechanisms of action and its target genes are largely unknown. In this study, we have combined RNA interference, in vitro DNA binding, chromatin immunoprecipitation sequencing and global gene-expression profiling to investigate the role of Sp2 for cellular functions, to define target sites and to identify genes regulated by Sp2. We show that Sp2 is important for cellular proliferation that it binds to GC-boxes and occupies proximal promoters of genes essential for vital cellular processes including gene expression, replication, metabolism and signalling. Moreover, we identified important key target genes and cellular pathways that are directly regulated by Sp2. Most significantly, Sp2 binds and activates numerous sequence-specific transcription factor and co-activator genes, and represses the whole battery of cholesterol synthesis genes. Our results establish Sp2 as a sequence-specific regulator of vitally important genes.


Subject(s)
Gene Expression Regulation , Sp2 Transcription Factor/metabolism , Animals , Base Sequence , Binding Sites , Cell Proliferation , DNA/chemistry , DNA/metabolism , Data Mining , Gene Deletion , Gene Expression Profiling , Genome , HEK293 Cells , HeLa Cells , Humans , Mice , Position-Specific Scoring Matrices , Promoter Regions, Genetic , RNA Interference , Sp1 Transcription Factor/metabolism , Sp2 Transcription Factor/antagonists & inhibitors , Sp2 Transcription Factor/genetics
11.
Exp Hematol ; 39(7): 730-40.e1-2, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21549805

ABSTRACT

OBJECTIVE: The transcription factor PAX5 is essential for the activation of B-cell-specific genes and for the silencing of myeloid-specific genes. We previously determined the molecular mechanism by which PAX5 silences the myeloid-specific colony-stimulating-factor-receptor (Csf1R) gene and showed that PAX5 directly binds to the Csf1r promoter as well as to an intronic enhancer that generates an antisense transcript in B cells. Here we examine the role of PAX5 in the regulation of sense and antisense transcription in B cells. MATERIALS AND METHODS: We performed PAX5-specific chromatin immunoprecipitation analyses across the Csfr1 locus. We investigated the role of PAX5 in regulating Csf1r sense and antisense promoter activity by transient transfections and by employing a Pax5(-/-) pro-B-cell line expressing an inducible PAX5 protein. PAX5 interacting factors were identified by pull-down experiments. The role of the transcription factor Sp3 in driving antisense promoter expression was examined in B cells from Sp3 knockout mice. RESULTS: PAX5 differentially regulates the Csf1r promoter and the promoter of the antisense transcript. PAX5 interferes with PU.1 transactivation at the sense promoter by binding to a PAX5 consensus sequence. At the antisense promoter, PAX5 does not specifically recognize DNA, but interacts with Sp3 to upregulate antisense promoter activity. Antisense promoter activation by PAX5 is dependent on the presence of its partial homeo-domain. CONCLUSIONS: We demonstrate that PAX5 regulates Csf1r in B cells by reducing the frequency of binding of the basal transcription machinery to the promoter and by activating antisense RNA expression.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , PAX5 Transcription Factor/genetics , Promoter Regions, Genetic/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , DNA, Antisense/genetics , Mice , Mice, Knockout , Mutation , PAX5 Transcription Factor/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
12.
PLoS Genet ; 6(11): e1001203, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085687

ABSTRACT

SUMO modification of transcription factors is linked to repression of transcription. The physiological significance of SUMO attachment to a particular transcriptional regulator, however, is largely unknown. We have employed the ubiquitously expressed murine transcription factor Sp3 to analyze the role of SUMOylation in vivo. We generated mice and mouse embryonic fibroblasts (MEFs) carrying a subtle point mutation in the SUMO attachment sequence of Sp3 (IKEE(553)D mutation). The E(553)D mutation impedes SUMOylation of Sp3 at K(551)in vivo, without affecting Sp3 protein levels. Expression profiling revealed that spermatocyte-specific genes, such as Dmc1 and Dnahc8, and neuronal genes, including Paqr6, Rims3, and Robo3, are de-repressed in non-testicular and extra-neuronal mouse tissues and in mouse embryonic fibroblasts expressing the SUMOylation-deficient Sp3E(553)D mutant protein. Chromatin immunoprecipitation experiments show that transcriptional de-repression of these genes is accompanied by the loss of repressive heterochromatic marks such as H3K9 and H4K20 tri-methylation and impaired recruitment of repressive chromatin-modifying enzymes. Finally, analysis of the DNA methylation state of the Dmc1, Paqr6, and Rims3 promoters by bisulfite sequencing revealed that these genes are highly methylated in Sp3wt MEFs but are unmethylated in Sp3E(553)D MEFs linking SUMOylation of Sp3 to tissue-specific CpG methylation. Our results establish SUMO conjugation to Sp3 as a molecular beacon for the assembly of repression machineries to maintain tissue-specific transcriptional gene silencing.


Subject(s)
Gene Silencing , Neurons/metabolism , Sp3 Transcription Factor/genetics , Spermatocytes/metabolism , Sumoylation/genetics , Animals , Chromatin/metabolism , DNA Methylation/genetics , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Targeting , Male , Meiosis/genetics , Mice , Mice, Mutant Strains , Neurons/cytology , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sp3 Transcription Factor/metabolism
13.
PLoS One ; 5(3): e9587, 2010 Mar 08.
Article in English | MEDLINE | ID: mdl-20221402

ABSTRACT

BACKGROUND: The zinc finger protein Sp2 (specificity protein 2) is a member of the glutamine-rich Sp family of transcription factors. Despite its close similarity to Sp1, Sp3 and Sp4, Sp2 does not bind to DNA or activate transcription when expressed in mammalian cell lines. The expression pattern and the biological relevance of Sp2 in the mouse are unknown. METHODOLOGY/PRINCIPAL FINDINGS: Whole-mount in situ hybridization of mouse embryos between E7.5 and E9.5 revealed abundant expression in most embryonic and extra-embryonic tissues. In order to unravel the biological relevance of Sp2, we have targeted the Sp2 gene by a tri-loxP strategy. Constitutive Sp2null and conditional Sp2cko knockout alleles were obtained by crossings with appropriate Cre recombinase expressing mice. Constitutive disruption of the mouse Sp2 gene (Sp2null) resulted in severe growth retardation and lethality before E9.5. Mouse embryonic fibroblasts (MEFs) derived from Sp2null embryos at E9.5 failed to grow. Cre-mediated ablation of Sp2 in Sp2cko/cko MEFs obtained from E13.5 strongly impaired cell proliferation. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that Sp2 is essential for early mouse development and autonomous proliferation of MEFs in culture. Comparison of the Sp2 knockout phenotype with the phenotypes of Sp1, Sp3 and Sp4 knockout strains shows that, despite their structural similarity and evolutionary relationship, all four glutamine-rich members of the Sp family of transcription factors have distinct non-redundant functions in vivo.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Sp2 Transcription Factor/physiology , Animals , Cell Proliferation , DNA/metabolism , Evolution, Molecular , Female , In Situ Hybridization , Male , Mice , Mice, Knockout , Phenotype , Time Factors , Transcription, Genetic
14.
Genes Dev ; 23(1): 118-32, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19136629

ABSTRACT

To elucidate the function of the transcriptional coregulator PRMT1 (protein arginine methyltranferase 1) in interferon (IFN) signaling, we investigated the expression of STAT1 (signal transducer and activator of transcription) target genes in PRMT1-depleted cells. We show here that PRMT1 represses a subset of IFNgamma-inducible STAT1 target genes in a methyltransferase-dependent manner. These genes are also regulated by the STAT1 inhibitor PIAS1 (protein inhibitor of activated STAT1). PIAS1 is arginine methylated by PRMT1 in vitro as well as in vivo upon IFN treatment. Mutational and mass spectrometric analysis of PIAS1 identifies Arg 303 as the single methylation site. Using both methylation-deficient and methylation-mimicking mutants, we find that arginine methylation of PIAS1 is essential for the repressive function of PRMT1 in IFN-dependent transcription and for the recruitment of PIAS1 to STAT1 target gene promoters in the late phase of the IFN response. Methylation-dependent promoter recruitment of PIAS1 results in the release of STAT1 and coincides with the decline of STAT1-activated transcription. Accordingly, knockdown of PRMT1 or PIAS1 enhances the anti-proliferative effect of IFNgamma. Our findings identify PRMT1 as a novel and crucial negative regulator of STAT1 activation that controls PIAS1-mediated repression by arginine methylation.


Subject(s)
Arginine/metabolism , Protein Inhibitors of Activated STAT/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Cell Line , Cell Proliferation , Gene Expression Regulation , Gene Knockdown Techniques , HeLa Cells , Humans , Interferon-gamma/metabolism , Interferons/metabolism , Methylation , Promoter Regions, Genetic
15.
EMBO Rep ; 9(9): 899-906, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18617891

ABSTRACT

Modification of many transcription factors including Sp3 and steroidogenic factor 1 with the small ubiquitin-like modifier (SUMO) is associated with transcriptional repression. Here, we show that SUMOylation of transcription factors bound to DNA provokes the establishment of compacted repressive chromatin with characteristics of heterochromatin. Chromatin immunoprecipitation experiments revealed SUMO-dependent recruitment of the chromatin remodeller Mi-2, MBT-domain proteins, heterochromatic protein 1, and the histone methyltransferases SETDB1 and SUV4-20H, concomitant with the establishment of histone modifications associated with repressed genes, including H3K9 and H4K20 trimethylation. These results indicate that SUMOylation has a crucial role in regulating gene expression by initiating chromatin structure changes that render DNA inaccessible to the transcription machinery.


Subject(s)
Gene Silencing , Heterochromatin/genetics , SUMO-1 Protein/metabolism , Sp3 Transcription Factor/metabolism , Transcription, Genetic/genetics , Animals , Cell Line , Chromatin Immunoprecipitation , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Histone-Lysine N-Methyltransferase , Humans , Mice , Models, Biological , Polymerase Chain Reaction/methods , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , SUMO-1 Protein/genetics , Sp3 Transcription Factor/genetics , Transfection
16.
Mol Cell ; 29(6): 742-54, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18374648

ABSTRACT

SUMO modification of many transcription factors is linked to transcriptional repression. The molecular mechanisms by which SUMO attachment represses transcription are largely unknown. Here we report a genome-wide RNA interference screen in Drosophila melanogaster cells for components regulating and mediating SUMO-dependent transcriptional repression. Analysis of >21,000 double-stranded RNAs (dsRNAs) identified 120 genes whose dsRNA-mediated knockdowns impaired SUMO-dependent transcriptional repression. Several of these genes encode chromatin-associated proteins, including the ATP-dependent chromatin remodeler Mi-2, the D. melanogaster ortholog of the C. elegans protein MEP-1, and the polycomb protein Sfmbt. Knockdown of these proteins did not impair SUMO conjugation, demonstrating that they act downstream of SUMO attachment. Biochemical analyses revealed that MEP-1, Mi-2, and Sfmbt interact with each other, bind to SUMO, and are recruited to promoters in a SUMOylation-dependent manner. Our results suggest that MEP-1, Mi-2, and Sfmbt are part of a common repression complex established by DNA-bound SUMO-modified transcription factors.


Subject(s)
Chromatin/genetics , Drosophila melanogaster/genetics , RNA Interference , SUMO-1 Protein/metabolism , Transcription, Genetic , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation , Genome , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Mammals , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Promoter Regions, Genetic , RNA, Double-Stranded/genetics , Species Specificity
17.
Mol Cell Biol ; 27(24): 8571-82, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17923686

ABSTRACT

Mice lacking the zinc finger transcription factor specificity protein 3 (Sp3) die prenatally in the C57BL/6 background. To elucidate the cause of mortality we analyzed the potential role of Sp3 in embryonic heart development. Sp3 null hearts display defective looping at embryonic day 10.5 (E10.5), and at E14.5 the Sp3 null mutants have developed a range of severe cardiac malformations. In an attempt to position Sp3 in the cardiac developmental hierarchy, we analyzed the expression patterns of >15 marker genes in Sp3 null hearts. Expression of cardiac ankyrin repeat protein (Carp) was downregulated prematurely after E12.5, while expression of the other marker genes was not affected. Chromatin immunoprecipitation analysis revealed that Sp3 is bound to the Carp promoter region in vivo. Microarray analysis indicates that small-molecule metabolism and cell-cell interactions are the most significantly affected biological processes in E12.5 Sp3 null myocardium. Since the epicardium showed distension from the myocardium, we studied expression of Wt1, a marker for epicardial cells. Wt1 expression was diminished in epicardium-derived cells in the myocardium of Sp3 null hearts. We conclude that Sp3 is required for normal cardiac development and suggest that it has a crucial role in myocardial differentiation.


Subject(s)
Heart Defects, Congenital/metabolism , Sp3 Transcription Factor/deficiency , Sp3 Transcription Factor/metabolism , Animals , Biomarkers/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/pathology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome , Heart/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins , Nuclear Proteins/genetics , Organ Size , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics , Sp3 Transcription Factor/genetics
18.
J Biol Chem ; 282(42): 31038-45, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17670746

ABSTRACT

Methylglyoxal is a highly reactive dicarbonyl degradation product formed from triose phosphates during glycolysis. Methylglyoxal forms stable adducts primarily with arginine residues of intracellular proteins. The biologic role of this covalent modification in regulating cell function is not known. Here we report that in mouse kidney endothelial cells, high glucose causes increased methylglyoxal modification of the corepressor mSin3A. Methylglyoxal modification of mSin3A results in increased recruitment of O-GlcNAc-transferase, with consequent increased modification of Sp3 by O-linked N-acetylglucosamine. This modification of Sp3 causes decreased binding to a glucose-responsive GC-box in the angiopoietin-2 (Ang-2) promoter, resulting in increased Ang-2 expression. Increased Ang-2 expression induced by high glucose increased expression of intracellular adhesion molecule 1 and vascular cell adhesion molecule 1 in cells and in kidneys from diabetic mice and sensitized microvascular endothelial cells to the proinflammatory effects of tumor necrosis factor alpha. This novel mechanism for regulating gene expression may play a role in the pathobiology of diabetic vascular disease.


Subject(s)
Angiopoietin-2/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Protein Processing, Post-Translational , Pyruvaldehyde/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Acetylglucosamine/genetics , Acetylglucosamine/metabolism , Angiopoietin-2/genetics , Animals , Arginine/genetics , Arginine/metabolism , Cell Line, Transformed , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Endothelial Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucose/pharmacology , Glycolysis/drug effects , Glycolysis/genetics , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Kidney/metabolism , Kidney/pathology , Mice , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Repressor Proteins/genetics , Response Elements/genetics , Sin3 Histone Deacetylase and Corepressor Complex , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sweetening Agents/metabolism , Sweetening Agents/pharmacology , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
19.
Dev Dyn ; 236(8): 2235-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17584888

ABSTRACT

The ubiquitously expressed zinc finger transcription factors Sp1 and Sp3 play critical roles in embryonic development. Sp1 knockout mice die around embryonic day 10.5. Mice lacking Sp3 are postnatal lethal. Mice heterozygous for either Sp1 or Sp3 are apparently normal, although slightly smaller. Here, we show that compound heterozygosity of Sp1 and Sp3 results in embryonic lethality accompanied by a spectrum of developmental abnormalities, including growth retardation, morphological alterations of the lung, impaired ossification, anemia, and placental defects. Anemia in Sp1/Sp3 compound heterozygous mutant embryos is associated with impaired maturation of erythrocytes. Analyses of the placenta revealed a markedly reduced spongiotrophoblast layer and a severe disorganization of the labyrinth layer in Sp1/Sp3 compound heterozygous as well as in Sp3-deficient mutant embryos. Our findings demonstrate that a threshold of Sp1 and Sp3 activity is required for normal embryonic development, suggesting that Sp1 and Sp3 act cooperatively to regulate downstream targets.


Subject(s)
Erythropoiesis/genetics , Placenta/abnormalities , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics , Animals , Heterozygote , Mice , Phenotype , Placenta/pathology , Sp1 Transcription Factor/deficiency , Sp3 Transcription Factor/deficiency , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...