Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 50(4): 2005-2018, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35137179

ABSTRACT

The second cell fate decision in the early stage of mammalian embryonic development is pivotal; however, the underlying molecular mechanism is largely unexplored. Here, we report that Prmt1 acts as an important regulator in primitive endoderm (PrE) formation. First, Prmt1 depletion promotes PrE gene expression in mouse embryonic stem cells (ESCs). Single-cell RNA sequencing and flow cytometry assays demonstrated that Prmt1 depletion in mESCs contributes to an emerging cluster, where PrE genes are upregulated significantly. Furthermore, the efficiency of extraembryonic endoderm stem cell induction increased in Prmt1-depleted ESCs. Second, the pluripotency factor Klf4 methylated at Arg396 by Prmt1 is required for recruitment of the repressive mSin3a/HDAC complex to silence PrE genes. Most importantly, an embryonic chimeric assay showed that Prmt1 inhibition and mutated Klf4 at Arg 396 induce the integration of mouse ESCs into the PrE lineage. Therefore, we reveal a regulatory mechanism for cell fate decisions centered on Prmt1-mediated Klf4 methylation.


Subject(s)
Embryo, Mammalian/metabolism , Endoderm , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cell Differentiation , Embryonic Development , Endoderm/metabolism , Female , Kruppel-Like Factor 4/metabolism , Mice , Mouse Embryonic Stem Cells , Pregnancy
2.
Stem Cell Res Ther ; 11(1): 195, 2020 05 24.
Article in English | MEDLINE | ID: mdl-32448390

ABSTRACT

Temperature is an important microenvironmental factor that functions epigenetically in normal embryonic development. However, the effect of hyperthermia in the stem cells is not fully understood. Oct4 is a tightly regulated master regulator of pluripotency maintenance in stem cells and during early embryonic development. We report here that Oct4 protein level was significantly reduced under hyperthermia in mouse blastocysts and embryonic stem cells. The reduction in Oct4 in the mouse embryonic stem cells under hyperthermia was mediated by a ubiquitin-proteasome pathway that was dependent on the activity of death-associated protein kinase 1 (Dapk1) to phosphorylate its substrate, Pin1. Our results imply that the depletion of Oct4 via brief hyperthermia, such as a high fever, during early pregnancy might severely impair the growth of the mammalian embryo or even cause its death.


Subject(s)
Gene Expression Regulation, Developmental , Hyperthermia , Animals , Blastocyst/metabolism , Embryonic Development , Embryonic Stem Cells/metabolism , Female , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy
3.
Biochim Biophys Acta Gene Regul Mech ; 1862(10): 194442, 2019 10.
Article in English | MEDLINE | ID: mdl-31634638

ABSTRACT

MyoD is a determining transcription factor involved in myogenic cell differentiation. Post translational modifications of MyoD, including phosphorylation and acetylation, can regulate its transcription activity. Inhibition of protein arginine methyltransferase 1 (PRMT1) leads to insufficient muscle differentiation. However, little is known about arginine methylation in regulating MyoD activity. Here, we demonstrated that MyoD interacts with PRMT1 via its bHLH domain. MyoD could be methylated by PRMT1 at R121. Moreover, R111 and R121 of MyoD are responsible for MyoD-mediated myogenin gene transcription in C2C12 cells. PRMT1 promotes MyoD-mediated myogenin expression, for which the enzymatic activity of PRMT1 is needed. The arginine methylation of MyoD by PRMT1 enhances its DNA binding activity and transactivation. Our data help to further clarify the molecular mechanism of PRMT1 in regulating muscle cell differentiation and provide a new therapeutic target for diseases caused by the abnormal differentiation of muscle cells.


Subject(s)
MyoD Protein/genetics , Myogenin/genetics , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Transcription, Genetic , Arginine/genetics , Cell Differentiation/genetics , Gene Expression Regulation/genetics , Humans , Methylation , Muscle Development/genetics , Protein Processing, Post-Translational/genetics
4.
Cancer Res ; 79(11): 2865-2877, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31015230

ABSTRACT

C/EBPα is an essential transcription factor involved in regulating the expression or function of certain cell-cycle regulators, including in breast cancer cells. Although protein arginine methyltransferases have been shown to play oncogenic roles in a variety of cancers, little is known about the role of arginine methylation in regulating the antiproliferation activity of C/EBPα. Here, we report that the protein arginine methyltransferase 1 (PRMT1) is overexpressed in human breast cancer and that elevated PRMT1 correlates with cancer malignancy. RNA-sequencing analysis revealed that knockdown of PRMT1 in breast cancer cells is accompanied by a decrease in the expression of pro-proliferative genes, including cyclin D1. Furthermore, tandem affinity purification followed by mass spectrometry identified PRMT1 as a component of the C/EBPα complex. C/EBPα associated with and was methylated by PRMT1 at three arginine residues (R35, R156, and R165). PRMT1-dependent methylation of C/EBPα promoted the expression of cyclin D1 by blocking the interaction between C/EBPα and its corepressor HDAC3, which resulted in rapid growth of tumor cells during the pathogenesis of breast cancer. Inhibition of PRMT1 significantly impeded the growth of cancer cells from patients with triple-negative breast cancer. This evidence that PRMT1 mediates C/EBPα methylation sheds light on a novel pathway and potential therapeutic target in breast cancer. SIGNIFICANCE: This study provides novel mechanistic insight of the role of the arginine methyltransferase PRMT1 in breast cancer pathogenesis.


Subject(s)
Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Animals , Arginine/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , CCAAT-Enhancer-Binding Proteins/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/metabolism , Humans , Methylation , Mice, Inbred BALB C , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Xenograft Model Antitumor Assays
5.
BMC Cancer ; 18(1): 27, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29301498

ABSTRACT

BACKGROUND: Lin28B and its paralog Lin28A are small RNA binding proteins that have similar inhibitory effects, although they target separate steps in the maturation of let-7 miRNAs in mammalian cells. Because Lin28B participates in the promotion and development of tumors mostly by blocking the let-7 tumor suppressor family members, we sought to explore the associated mechanisms to gain insights into how Lin28B might be decreased in human cancer cells to increase let-7 levels and reverse malignancy. RESULTS: We demonstrated that the histone acetyltransferase PCAF, via its cold shock domain, directly interacts with and subsequently acetylates Lin28B in lung adenocarcinoma-derived H1299 cells. RT-qPCR assays showed that both let-7a-1 and let-7g were increased in PCAF-transfected H1299 cells. Lin28B is acetylated by ectopic PCAF and translocates from the nucleus to the cytoplasm in H1299 cells. CONCLUSIONS: The effects of acetylated Lin28B on let-7a-1 and let-7g are similar to that of stable knockdown of Lin28B in H1299 cells. The new role of PCAF in mediating Lin28B acetylation and the specific release of its target microRNAs in H1299 cells may shed light on the potential application of let-7 in the clinical treatment of lung cancer patients.


Subject(s)
Adenocarcinoma/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , p300-CBP Transcription Factors/genetics , Acetylation , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Protein Binding
6.
Biochim Biophys Acta Gene Regul Mech ; 1860(4): 516-522, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28235567

ABSTRACT

CR6-interacting factor 1 (CRIF1) is ubiquitously expressed in human tissues. CRIF1 was first identified as a Gadd45γ (also known as CR6)-interacting protein, and it was also identified in a human colon cancer cell line stably transformed with p53. These results suggested that CRIF1 functions in the nucleus with p53 and Gadd45 family proteins in the suppression of cell growth and tumor development. Here, we found that CRIF1 could be recruited to a specific region in the promoter of the p53 gene, eliciting an increase in the mRNA and protein levels of p53 as well as p53 functional target genes. These functions required CRIF1 to interact with SNF5. CRIF1 was further recruited to the upstream promoter region of the p53 gene to suppress cell cycle progression in HCT116 cells. To our knowledge, this is the first evidence indicating that SNF5 is indispensable for CRIF1-enhanced p53 activity and its function in the suppression of cell cycle arrest in human cancer cells.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Nuclear Proteins/metabolism , SMARCB1 Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Cycle Checkpoints/genetics , G1 Phase/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , Protein Binding/genetics , S Phase/genetics , Transcriptional Activation/genetics
7.
Biochim Biophys Acta ; 1859(2): 348-54, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26704017

ABSTRACT

Mammalian Sirtuin proteins (SIRTs) are homologs of yeast Sir2, and characterized as class III histone deacetylases of NAD(+) dependence. Unlike their lower counterparts that are directly involved in the extending of lifespan, mammalian SIRTs mainly function in metabolism and cellular homeostasis, among them, SIRT7 is the least understood. SIRT7 is localized in the nucleus and rich in nucleoli associated with RNA polymerase I, and correlated with cell proliferation. In contrast, SIRT7 has recently been demonstrated to specifically deacetylate H3K18ac in the chromatin, and in most cases represses proliferation. Although MicroRNA as miR-125b has been reported to down-regulate SIRT7 by binding to its 3'UTR, however, how SIRT7 gene is regulated remains unclear. Here, we identified the transcription initiation site of human SIRT7 gene at the upstream 23rd A nucleotide respective to the translational codon, and the SIRT7 is a TATA-less and initiator-less gene. The sequences in the upstream region between -256 and -129 bp are identical with important functions in the three species detected. A C/EBPα responding element is found that binds both C/EBPα and C/EBPß in vitro. We showed TSA induced SIRT7 gene transcription and only the HDAC3, but not its catalytic domain depleted mutant, interacted with C/EBPα to occupy the C/EBPα element and repressed SIRT7 gene in the hepatocellular carcinoma cells. To our knowledge, this is the first report on the regulation mechanism of SIRT7 gene, in which, HDAC3 collaborated with C/EBPα to occupy its responding element in the upstream region of SIRT7 gene and repressed its expression in human cells.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Carcinoma, Hepatocellular/genetics , Histone Deacetylases/genetics , Liver Neoplasms/genetics , Sirtuins/genetics , 3' Untranslated Regions , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/biosynthesis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chromatin/genetics , Histone Deacetylases/biosynthesis , Humans , Liver Neoplasms/pathology , Promoter Regions, Genetic , Sirtuins/biosynthesis
8.
PLoS Biol ; 12(12): e1002026, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25535969

ABSTRACT

Histone lysine (K) residues, which are modified by methyl- and acetyl-transferases, diversely regulate RNA synthesis. Unlike the ubiquitously activating effect of histone K acetylation, the effects of histone K methylation vary with the number of methyl groups added and with the position of these groups in the histone tails. Histone K demethylases (KDMs) counteract the activity of methyl-transferases and remove methyl group(s) from specific K residues in histones. KDM3A (also known as JHDM2A or JMJD1A) is an H3K9me2/1 demethylase. KDM3A performs diverse functions via the regulation of its associated genes, which are involved in spermatogenesis, metabolism, and cell differentiation. However, the mechanism by which the activity of KDM3A is regulated is largely unknown. Here, we demonstrated that mitogen- and stress-activated protein kinase 1 (MSK1) specifically phosphorylates KDM3A at Ser264 (p-KDM3A), which is enriched in the regulatory regions of gene loci in the human genome. p-KDM3A directly interacts with and is recruited by the transcription factor Stat1 to activate p-KDM3A target genes under heat shock conditions. The demethylation of H3K9me2 at the Stat1 binding site specifically depends on the co-expression of p-KDM3A in the heat-shocked cells. In contrast to heat shock, IFN-γ treatment does not phosphorylate KDM3A via MSK1, thereby abrogating its downstream effects. To our knowledge, this is the first evidence that a KDM can be modified via phosphorylation to determine its specific binding to target genes in response to thermal stress.


Subject(s)
Gene Expression Regulation , Heat-Shock Response/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Amino Acid Sequence , Base Sequence , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation/drug effects , Genome, Human , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Humans , Interferon-gamma/pharmacology , Jumonji Domain-Containing Histone Demethylases/chemistry , Jurkat Cells , Models, Biological , Molecular Sequence Data , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stress, Physiological/drug effects , Stress, Physiological/genetics
9.
J Mol Cell Biol ; 4(4): 231-41, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22368283

ABSTRACT

Ribosome biogenesis is critical in the growth of eukaryotic cells, in which the synthesis of precursor ribosomal RNA is the first and rate-limiting step. Here, we show that human PIH1 domain-containing protein 1 (PIH1) interacts directly with histone H4 and recruits the Brg1-SWI/SNF complex via SNF5 to human rRNA genes. This process is likely involved in PIH1-dependent DNase I-hypersensitive chromatin remodeling at the core promoter of the rRNA genes. PIH1 mediates the occupancy of not only the Brg1 complex but also the Pol I complex at the core promoter and enhances transcription initiation of rRNA genes. Additionally, the interaction between PIH1 and H4K16 expels TIP5, a component of the silencing nucleolar remodeling complex (NoRC), from the core region, suggesting that PIH1 is involved in the derepression of NoRC-silenced rRNA genes. These data indicate that PIH1 is a positive regulator of human rRNA genes and is of great importance for the recovery of human cells from nutrient starvation and the transition to glucose-induced exponential growth in vivo.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Glucose/pharmacology , Histones/metabolism , RNA Precursors/biosynthesis , Chromatin Assembly and Disassembly/drug effects , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins , Deoxyribonuclease I/metabolism , Gene Expression Regulation/drug effects , Genes, rRNA/genetics , HEK293 Cells , Humans , Models, Biological , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , SMARCB1 Protein , Transcription Factors
10.
Cell Res ; 20(12): 1345-60, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21079652

ABSTRACT

The SWI/SNF chromatin-remodeling complexes utilize energy from ATP hydrolysis to reposition nucleosomes and regulate the expression of human genes. Here, we studied the roles of human Brahma (hBrm) and Brahma-related gene 1 (Brg1), the ATPase subunits of the SWI/SNF complexes, in regulating human genes. Our results indicate that both hBrm and Brg1 interact with Signal transducer and activator of transcription (Stat) 1 in vitro. However, Stat1 in its native form only recruits hBrm to IFNγ-activated sequences (GAS) of individual genes; by contrast, in a stress-induced phosphorylated form, Stat1 mainly binds to Brg1. Under basal conditions, hBrm is recruited by native Stat1 to the GAS and exists in a mSin3/HDAC co-repressor complex on the hsp90α gene, which shows a compact chromatin structure. Upon heat-shock, hBrm is acetylated by p300 and dissociates from the co-repressor complex, which the phosphorylated Stat1 is increased, and binds and recruits Brg1 to the GAS, leading to elevated induction of the gene. This hBrm/Brg1 switch also occurs at the GAS of all of the three examined immune genes in heat-shocked cells; however, this switch only occurs in specific cell types upon exposure to IFNγ. Regardless of the stimulus, the hBrm/Brg1 switch at the GAS elicits an increase in gene activity. Our data are consistent with the hypothesis that the hBrm/Brg1 switch is an indicator of the responsiveness of a gene to heat-shock or IFNγ stimulation and may represent an "on-off switch" of gene expression in vivo.


Subject(s)
DNA Helicases/metabolism , Interferon-gamma/pharmacology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Acetylation , Amino Acid Sequence , Chromatin Assembly and Disassembly , DNA Helicases/genetics , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Phosphorylation , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Temperature , Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
11.
Cell Signal ; 22(8): 1206-13, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20353823

ABSTRACT

We have reported earlier that a heat shock element in the first intron of human hsp90beta gene (iHSE) acts as an intronic enhancer to bind the heat shock factor (HSF1) and activates hsp90beta gene under heat shock. Here, we show that, in addition to the HSF1, Stat1 phosphorylation is indispensable in the event. We show that Jak2, a Janus kinase specifically associated with the beta subunit of IFNgamma receptor, and PKCepsilon an isoform of the atypical PKC family, are the two dominant kinases responsible for the heat shock induced phosphorylation on Y701 and S727 of Stat1. However, the activation of these kinases under heat shock requires the association of chaperone proteins of the Hsp90 family, in particular, the Hsp90beta under heat shock. Furthermore, Brg1, an ATPase subunit of the SWI/SNF chromatin remodeling complex is likely recruited by HSF1 and Stat1 at the iHSE under heat shock. Brg1 further confers an open chromatin conformation at the promoter region that is pivotal to the heat shock induced fully activation of the hsp90beta gene in Jurkat cells. This is a novel example of how multiple activation steps occur under heat shock, first on the kinases and then the Stat1 and the SWI/SNF chromatin remodeling complex that follows to conduct an auto-regulation based fully activation of the gene.


Subject(s)
HSP90 Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , STAT1 Transcription Factor/metabolism , Benzoquinones/pharmacology , DNA Helicases/metabolism , Enhancer Elements, Genetic , Homeostasis , Humans , Introns , Janus Kinase 2/metabolism , Jurkat Cells , Lactams, Macrocyclic/pharmacology , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Kinase C-epsilon/metabolism , Transcription Factors/metabolism
12.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 31(6): 746-50, 2009 Dec.
Article in Chinese | MEDLINE | ID: mdl-20078946

ABSTRACT

OBJECTIVE: To explore the effect of a non-lethal heat shock, in comparison with the treatment of interferon-gamma (IFN gamma), on the expression of major histocompatibility complex transactivator (CTA) and its downstream target gene of the human leukocyte antigens (HLA)-DR in Jurkat cells. METHODS: The changes of CTA mRNA in Jurkat cells before and after the treatment of heat shock or IFN gamma were detected using real time RT-PCR. The changes of CTA protein were detected with Western blot. The expression of HLA-DR was detected with flow cytometry. : CTA mRNA and protein were induced in Jurkat cells under heat shock, but not with IFN-gamma. The expression of HLA-DR gene significantly increased after recovery (P<0.01). CONCLUSION: The expressions of CTA and HLA-DR in Jurkat cells remarkably increase after heat shock, indicating that heat shock may help reconstruct relevant genes in cells with immunologic gene deficiencies.


Subject(s)
HLA-DR Antigens/metabolism , Heat-Shock Response/physiology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Humans , Jurkat Cells , Nuclear Proteins/genetics , RNA, Messenger/genetics , Trans-Activators/genetics
13.
J Biol Chem ; 282(26): 18872-8, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17468105

ABSTRACT

Myogenin and its upstream regulator MyoD are known to be required for myogenic cell differentiation. Although both of them can be expressed in rhabdomyosarcoma-derived RD cells, the cells are unable to undergo full-scale terminal myogenic differentiation. 12-O-Tetradecanoylphorbol-13-acetate (TPA) has been found to be functional in the induction of RD cell differentiation, whereas its mechanism is not fully understood. By using quantitative real-time-based chromatin immunoprecipitation and real-time reverse transcription-PCR-based promoter activity assays, we examined the activation mechanism of the myogenin gene during TPA-induced differentiation of the RD cells. We have shown that a histone acetyltransferase PCAF and ATPase subunit BRG1 of the SWI/SNF chromatin remodeling complex are sequentially recruited to the promoter of the myogenin gene. Both PCAF and BRG1 are also involved in the activation of the myogenin gene. In addition, we have found that the p38 mitogen-activated protein kinase is required for BRG1 recruitment in TPA-mediated myogenin induction. We propose that there are two distinct activation steps for the induction of myogenin in TPA-induced early differentiation of RD cells: 1) an early step that requires PCAF activity to acetylate core histones and MyoD to initiate myogenin gene expression, and 2) a later step that requires p38-dependent activity of the SWI/SNF remodeling complex to provide an open conformation for the induction of myogenin. Our studies reveal an essential role for epigenetic regulation in TPA-induced differentiation of RD cells and provide potential drug targets for future treatment of the rhabdomyosarcoma.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Helicases/metabolism , Histone Acetyltransferases/metabolism , Myogenin/metabolism , Nuclear Proteins/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Transcription Factors/metabolism , Acetylation , Carcinogens/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Chromatin/physiology , Drug Design , Epigenesis, Genetic/physiology , Gene Expression Regulation, Neoplastic , Humans , Myogenin/genetics , Promoter Regions, Genetic/physiology , Rhabdomyosarcoma/drug therapy , Tetradecanoylphorbol Acetate/pharmacology , p300-CBP Transcription Factors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...