Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Nat Commun ; 15(1): 2809, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561334

ABSTRACT

Protein arginine methyltransferase 9 (PRMT9) is a recently identified member of the PRMT family, yet its biological function remains largely unknown. Here, by characterizing an intellectual disability associated PRMT9 mutation (G189R) and establishing a Prmt9 conditional knockout (cKO) mouse model, we uncover an important function of PRMT9 in neuronal development. The G189R mutation abolishes PRMT9 methyltransferase activity and reduces its protein stability. Knockout of Prmt9 in hippocampal neurons causes alternative splicing of ~1900 genes, which likely accounts for the aberrant synapse development and impaired learning and memory in the Prmt9 cKO mice. Mechanistically, we discover a methylation-sensitive protein-RNA interaction between the arginine 508 (R508) of the splicing factor 3B subunit 2 (SF3B2), the site that is exclusively methylated by PRMT9, and the pre-mRNA anchoring site, a cis-regulatory element that is critical for RNA splicing. Additionally, using human and mouse cell lines, as well as an SF3B2 arginine methylation-deficient mouse model, we provide strong evidence that SF3B2 is the primary methylation substrate of PRMT9, thus highlighting the conserved function of the PRMT9/SF3B2 axis in regulating pre-mRNA splicing.


Subject(s)
Alternative Splicing , RNA , Animals , Humans , Mice , Arginine/metabolism , Mice, Knockout , Mutation , Protein-Arginine N-Methyltransferases/metabolism , RNA/metabolism , RNA Precursors/metabolism , RNA Splicing/genetics
2.
Leukemia ; 38(6): 1236-1245, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643304

ABSTRACT

Targeting the metabolic dependencies of acute myeloid leukemia (AML) cells is a promising therapeutical strategy. In particular, the cysteine and methionine metabolism pathway (C/M) is significantly altered in AML cells compared to healthy blood cells. Moreover, methionine has been identified as one of the dominant amino acid dependencies of AML cells. Through RNA-seq, we found that the two nucleoside analogs 8-chloro-adenosine (8CA) and 8-amino-adenosine (8AA) significantly suppress the C/M pathway in AML cells, and methionine-adenosyltransferase-2A (MAT2A) is one of most significantly downregulated genes. Additionally, mass spectrometry analysis revealed that Venetoclax (VEN), a BCL-2 inhibitor recently approved by the FDA for AML treatment, significantly decreases the intracellular level of methionine in AML cells. Based on these findings, we hypothesized that combining 8CA or 8AA with VEN can efficiently target the Methionine-MAT2A-S-adenosyl-methionine (SAM) axis in AML. Our results demonstrate that VEN and 8CA/8AA synergistically decrease the SAM biosynthesis and effectively target AML cells both in vivo and in vitro. These findings suggest the promising potential of combining 8CA/8AA and VEN for AML treatment by inhibiting Methionine-MAT2A-SAM axis and provide a strong rationale for our recently activated clinical trial.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Drug Synergism , Leukemia, Myeloid, Acute , Methionine Adenosyltransferase , Methionine , S-Adenosylmethionine , Sulfonamides , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Humans , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Sulfonamides/pharmacology , Methionine/metabolism , Methionine/analogs & derivatives , Methionine Adenosyltransferase/metabolism , Methionine Adenosyltransferase/antagonists & inhibitors , Methionine Adenosyltransferase/genetics , Animals , Mice , S-Adenosylmethionine/pharmacology , S-Adenosylmethionine/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Xenograft Model Antitumor Assays , Cell Line, Tumor
3.
J Med Chem ; 66(19): 13665-13683, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37560786

ABSTRACT

Less studied than the other protein arginine methyltransferase isoforms, PRMT7 and PRMT9 have recently been identified as important therapeutic targets. Yet, most of their biological roles and functions are still to be defined, as well as the structural requirements that could drive the identification of selective modulators of their activity. We recently described the structural requirements that led to the identification of potent and selective PRMT4 inhibitors spanning both the substrate and the cosubstrate pockets. The reanalysis of the data suggested a PRMT7 preferential binding for shorter derivatives and prompted us to extend these structural studies to PRMT9. Here, we report the identification of the first potent PRMT7/9 inhibitor and its binding mode to the two PRMT enzymes. Label-free quantification mass spectrometry confirmed significant inhibition of PRMT activity in cells. We also report the setup of an effective AlphaLISA assay to screen small molecule inhibitors of PRMT9.


Subject(s)
Protein-Arginine N-Methyltransferases , Arginine/chemistry , Methylation , Protein-Arginine N-Methyltransferases/antagonists & inhibitors
4.
Methods Mol Biol ; 2666: 265-278, 2023.
Article in English | MEDLINE | ID: mdl-37166671

ABSTRACT

R-loops are three-stranded nucleic acid structures that consist of a DNA-RNA hybrid and a displaced single-stranded DNA. Since it was first reported by Ronald Davis and colleagues over 40 years ago, the study of R-loops has become an increasingly expanded area of research. Numerous factors have been identified to modulate the dynamic formation and resolution of R-loops, which are critical for proper controls of gene expression and genome stability. Along the lines of these discoveries, various biochemical and cellular assays have been developed to detect R-loop changes in vitro and in vivo. In this chapter, we describe a protocol for measuring R-loop formation using a plasmid-based in vitro transcription assay. The R-loop formed is then detected and quantified by using gel mobility, antibody staining, and DNA-RNA immunoprecipitation (DRIP)-qPCR assays. Unlike the helicase assay that uses short R-loop substrates, this assay system introduces DNA topology and active transcription as additional variables that impact R-loop formation, thus, more closely recapitulating in vivo situations. Furthermore, this method can be adopted for investigation of cis-elements and trans-acting factors that influence R-loop formation.


Subject(s)
DNA , R-Loop Structures , DNA/chemistry , RNA/chemistry , Plasmids/genetics , DNA, Single-Stranded
5.
Curr Opin Oncol ; 34(5): 546-551, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35788128

ABSTRACT

PURPOSE OF REVIEW: In this review, we summarize the biological roles of methionine, methionine adenosyl transferase 2A (MAT2A) and S -adenosyl methionine (SAM) in methylation reactions during tumorigenesis. Newly emerged inhibitors targeting the methionine-MAT2A-SAM axis will be discussed. RECENT FINDINGS: SAM is the critical and global methyl-donor for methylation reactions regulating gene expression, and in mammalian cells, it is synthesized by MAT2A using methionine. Recent studies have validated methionine and MAT2A as metabolic dependencies of cancer cells because of their essential roles in SAM biosynthesis. MAT2A inhibition leads to synthetic lethality in methylthioadenosine-phosphorylase (MTAP)-deleted cancers, which accounts for 15% of all cancer types. Of note, remarkable progress has been made in developing inhibitors targeting the methionine-MAT2A-SAM axis, as the first-in-class MAT2A inhibitors AG-270 and IDE397 enter clinical trials to treat cancer. SUMMARY: The methionine-MAT2A-SAM axis plays an important role in tumorigenesis by providing SAM as a critical substrate for abnormal protein as well as DNA and RNA methylation in cancer cells. Targeting SAM biosynthesis through MAT2A inhibition has emerged as a novel and promising strategy for cancer therapy.


Subject(s)
Neoplasms , Animals , Carcinogenesis , Humans , Mammals/metabolism , Methionine/metabolism , Methionine Adenosyltransferase/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , S-Adenosylmethionine/metabolism
7.
Nucleic Acids Res ; 49(15): 8573-8591, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34329467

ABSTRACT

R-loops, which consist of a DNA/RNA hybrid and a displaced single-stranded DNA (ssDNA), are increasingly recognized as critical regulators of chromatin biology. R-loops are particularly enriched at gene promoters, where they play important roles in regulating gene expression. However, the molecular mechanisms that control promoter-associated R-loops remain unclear. The epigenetic 'reader' Tudor domain-containing protein 3 (TDRD3), which recognizes methylarginine marks on histones and on the C-terminal domain of RNA polymerase II, was previously shown to recruit DNA topoisomerase 3B (TOP3B) to relax negatively supercoiled DNA and prevent R-loop formation. Here, we further characterize the function of TDRD3 in R-loop metabolism and introduce the DExH-box helicase 9 (DHX9) as a novel interaction partner of the TDRD3/TOP3B complex. TDRD3 directly interacts with DHX9 via its Tudor domain. This interaction is important for recruiting DHX9 to target gene promoters, where it resolves R-loops in a helicase activity-dependent manner to facilitate gene expression. Additionally, TDRD3 also stimulates the helicase activity of DHX9. This stimulation relies on the OB-fold of TDRD3, which likely binds the ssDNA in the R-loop structure. Thus, DHX9 functions together with TOP3B to suppress promoter-associated R-loops. Collectively, these findings reveal new functions of TDRD3 and provide important mechanistic insights into the regulation of R-loop metabolism.


Subject(s)
DEAD-box RNA Helicases/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Proteins/metabolism , R-Loop Structures , Chromatin , DNA Topoisomerases, Type I/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Protein Interaction Domains and Motifs , Proteins/chemistry , Transcription, Genetic
8.
EMBO J ; 40(5): e106309, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33459381

ABSTRACT

The N6-methyladenosine (m6 A) RNA modification serves crucial functions in RNA metabolism; however, the molecular mechanisms underlying the regulation of m6 A are not well understood. Here, we establish arginine methylation of METTL14, a component of the m6 A methyltransferase complex, as a novel pathway that controls m6 A deposition in mammalian cells. Specifically, protein arginine methyltransferase 1 (PRMT1) interacts with, and methylates the intrinsically disordered C terminus of METTL14, which promotes its interaction with RNA substrates, enhances its RNA methylation activity, and is crucial for its interaction with RNA polymerase II (RNAPII). Mouse embryonic stem cells (mESCs) expressing arginine methylation-deficient METTL14 exhibit significantly reduced global m6 A levels. Transcriptome-wide m6 A analysis identified 1,701 METTL14 arginine methylation-dependent m6 A sites located in 1,290 genes involved in various cellular processes, including stem cell maintenance and DNA repair. These arginine methylation-dependent m6 A sites are associated with enhanced translation of genes essential for the repair of DNA interstrand crosslinks; thus, METTL14 arginine methylation-deficient mESCs are hypersensitive to DNA crosslinking agents. Collectively, these findings reveal important aspects of m6 A regulation and new functions of arginine methylation in RNA metabolism.


Subject(s)
Adenosine/analogs & derivatives , Arginine/chemistry , Methyltransferases/metabolism , Mouse Embryonic Stem Cells/metabolism , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , RNA Polymerase II/metabolism , Adenosine/chemistry , Animals , Cytoplasm , Methyltransferases/chemistry , Methyltransferases/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Protein-Arginine N-Methyltransferases/genetics , RNA Polymerase II/genetics , Transcriptome
9.
Neural Plast ; 2020: 6137083, 2020.
Article in English | MEDLINE | ID: mdl-32684921

ABSTRACT

Hereditary hearing loss is one of the most common sensory disabilities worldwide. Mutation of POU domain class 4 transcription factor 3 (POU4F3) is considered the pathogenic cause of autosomal dominant nonsyndromic hearing loss (ADNSHL), designated as autosomal dominant nonsyndromic deafness 15. In this study, four novel variants in POU4F3, c.696G>T (p.Glu232Asp), c.325C>T (p.His109Tyr), c.635T>C (p.Leu212Pro), and c.183delG (p.Ala62Argfs∗22), were identified in four different Chinese families with ADNSHL by targeted next-generation sequencing and Sanger sequencing. Based on the American College of Medical Genetics and Genomics guidelines, c.183delG (p.Ala62Argfs∗22) is classified as a pathogenic variant, c.696G>T (p.Glu232Asp) and c.635T>C (p.Leu212Pro) are classified as likely pathogenic variants, and c.325C>T (p.His109Tyr) is classified as a variant of uncertain significance. Based on previous reports and the results of this study, we speculated that POU4F3 pathogenic variants are significant contributors to ADNSHL in the East Asian population. Therefore, screening of POU4F3 should be a routine examination for the diagnosis of hereditary hearing loss.


Subject(s)
Hearing Loss, Sensorineural/genetics , Homeodomain Proteins/genetics , Mutation, Missense , Pedigree , Transcription Factor Brn-3C/genetics , Adolescent , Child , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Young Adult
10.
Br J Cancer ; 122(9): 1288-1297, 2020 04.
Article in English | MEDLINE | ID: mdl-32147668

ABSTRACT

BACKGROUND: Previous studies suggested that mdivi-1 (mitochondrial division inhibitor), a putative inhibitor of dynamin-related protein (DRP1), decreased cancer cell proliferation through inducing mitochondrial fusion and altering oxygen consumption. However, the metabolic reprogramming underlying the DRP1 inhibition is still unclear in cancer cells. METHODS: To better understand the metabolic effect of DRP1 inhibition, [U-13C]glucose isotope tracing was employed to assess mdivi-1 effects in several cancer cell lines, DRP1-WT (wild-type) and DRP1-KO (knockout) H460 lung cancer cells and mouse embryonic fibroblasts (MEFs). RESULTS: Mitochondrial staining confirmed that mdivi-1 treatment and DRP1 deficiency induced mitochondrial fusion. Surprisingly, metabolic isotope tracing found that mdivi-1 decreased mitochondrial oxidative metabolism in the lung cancer cell lines H460, A549 and the colon cancer cell line HCT116. [U-13C]glucose tracing studies also showed that the TCA cycle intermediates had significantly lower enrichment in mdivi-1-treated cells. In comparison, DRP1-WT and DRP1-KO H460 cells had similar oxidative metabolism, which was decreased by mdivi-1 treatment. Furthermore, mdivi-1-mediated effects on oxidative metabolism were independent of mitochondrial fusion. CONCLUSIONS: Our data suggest that, in cancer cells, mdivi-1, a putative inhibitor of DRP1, decreases oxidative metabolism to impair cell proliferation.


Subject(s)
Dynamins/genetics , Mitochondria/drug effects , Oxidative Stress/drug effects , Quinazolinones/pharmacology , A549 Cells , Animals , Carbon Isotopes/chemistry , Carbon Isotopes/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dynamins/antagonists & inhibitors , Gene Knockout Techniques , Glucose/chemistry , Glucose/pharmacology , HCT116 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Oxygen Consumption/drug effects
11.
Blood ; 134(15): 1257-1268, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31395602

ABSTRACT

Relapse remains the main cause of MLL-rearranged (MLL-r) acute lymphoblastic leukemia (ALL) treatment failure resulting from persistence of drug-resistant clones after conventional chemotherapy treatment or targeted therapy. Thus, defining mechanisms underlying MLL-r ALL maintenance is critical for developing effective therapy. PRMT1, which deposits an asymmetric dimethylarginine mark on histone/non-histone proteins, is reportedly overexpressed in various cancers. Here, we demonstrate elevated PRMT1 levels in MLL-r ALL cells and show that inhibition of PRMT1 significantly suppresses leukemic cell growth and survival. Mechanistically, we reveal that PRMT1 methylates Fms-like receptor tyrosine kinase 3 (FLT3) at arginine (R) residues 972 and 973 (R972/973), and its oncogenic function in MLL-r ALL cells is FLT3 methylation dependent. Both biochemistry and computational analysis demonstrate that R972/973 methylation could facilitate recruitment of adaptor proteins to FLT3 in a phospho-tyrosine (Y) residue 969 (Y969) dependent or independent manner. Cells expressing R972/973 methylation-deficient FLT3 exhibited more robust apoptosis and growth inhibition than did Y969 phosphorylation-deficient FLT3-transduced cells. We also show that the capacity of the type I PRMT inhibitor MS023 to inhibit leukemia cell viability parallels baseline FLT3 R972/973 methylation levels. Finally, combining FLT3 tyrosine kinase inhibitor PKC412 with MS023 treatment enhanced elimination of MLL-r ALL cells relative to PKC412 treatment alone in patient-derived mouse xenografts. These results indicate that abolishing FLT3 arginine methylation through PRMT1 inhibition represents a promising strategy to target MLL-r ALL cells.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Apoptosis , Cell Proliferation , Cell Survival , Gene Rearrangement , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Tumor Cells, Cultured
12.
Cancer Res ; 79(1): 21-32, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30352814

ABSTRACT

Protein arginine methyltransferases (PRMT) are generally not mutated in diseased states, but they are overexpressed in a number of cancers, including breast cancer. To address the possible roles of PRMT overexpression in mammary gland tumorigenesis, we generated Cre-activated PRMT1, CARM1, and PRMT6 overexpression mouse models. These three enzymes are the primary type I PRMTs and are responsible for the majority of the asymmetric arginine methylation deposited in the cells. Using either a keratin 5-Cre recombinase (K5-Cre) cross or an MMTV-NIC mouse, we investigated the impact of PRMT overexpression alone or in the context of a HER2-driven model of breast cancer, respectively. The overexpression of all three PRMTs induced hyper-branching of the mammary glands and increased Ki-67 staining. When combined with the MMTV-NIC model, these in vivo experiments provided the first genetic evidence implicating elevated levels of these three PRMTs in mammary gland tumorigenesis, albeit with variable degrees of tumor promotion and latency. In addition, these mouse models provided valuable tools for exploring the biological roles and molecular mechanisms of PRMT overexpression in the mammary gland. For example, transcriptome analysis of purified mammary epithelial cells isolated from bigenic NIC-PRMT1 Tg and NIC-PRMT6 Tg mice revealed a deregulated PI3K-AKT pathway. In the future, these PRMT Tg lines can be leveraged to investigate the roles of arginine methylation in other tissues and tumor model systems using different tissue-specific Cre crosses, and they can also be used for testing the in vivo efficacy of small molecule inhibitors that target these PRMT. SIGNIFICANCE: These findings establish Cre-activated mouse models of three different arginine methyltransferases, PRMT1, CARM1, and PRMT6, which are overexpressed in human cancers, providing a valuable tool for the study of PRMT function in tumorigenesis.See related commentary by Watson and Bitler, p. 3.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Nuclear Proteins/physiology , Oncogenes , Protein-Arginine N-Methyltransferases/physiology , Repressor Proteins/physiology , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Transgenic , Signal Transduction
13.
Life Sci Alliance ; 1(5): e201800117, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30456381

ABSTRACT

The coactivator-associated arginine methyltransferase (CARM1) functions as a regulator of transcription by methylating a diverse array of substrates. To broaden our understanding of CARM1's mechanistic actions, we sought to identify additional substrates for this enzyme. To do this, we generated CARM1 substrate motif antibodies, and used immunoprecipitation coupled with mass spectrometry to identify cellular targets of CARM1, including mediator complex subunit 12 (MED12) and the lysine methyltransferase KMT2D. Both of these proteins are implicated in enhancer function. We identified the major CARM1-mediated MED12 methylation site as arginine 1899 (R1899), which interacts with the Tudor domain-containing effector molecule, TDRD3. Chromatin immunoprecipitation-seq studies revealed that CARM1 and the methyl mark it deposits are tightly associated with ERα-specific enhancers and positively modulate transcription of estrogen-regulated genes. In addition, we showed that the methylation of MED12, at the R1899 site, and the recruitment of TDRD3 by this methylated motif are critical for the ability of MED12 to interact with activating noncoding RNAs.

14.
J Biol Chem ; 293(39): 15290-15303, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30131339

ABSTRACT

Glucose is a critical nutrient for cell proliferation. However, the molecular pathways that regulate glucose metabolism are still elusive. We discovered that co-activator-associated arginine methyltransferase 1 (CARM1) suppresses glucose metabolism toward serine biosynthesis. By tracing the 13C-labeled glucose, we found that Carm1 knockout mouse embryonic fibroblasts exhibit significantly increased de novo serine synthesis than WT cells. This is caused, at least in part, by the reduced pyruvate kinase (PK) activity in these cells. The M2 isoform of PK (PKM2) is arginine-methylated by CARM1, and methylation enhances its activity. Mechanistically, CARM1 methylates PKM2 at arginines 445 and 447, which enhances PKM2 tetramer formation. Consequently, Carm1 knockout cells exhibit significant survival advantages over WT cells when extracellular serine is limited, likely due to their enhanced de novo serine synthesis capacity. Altogether, we identified CARM1 as an important regulator of glucose metabolism and serine synthesis.


Subject(s)
Carrier Proteins/genetics , Glucose/genetics , Membrane Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics , Pyruvate Kinase/genetics , Serine/biosynthesis , Thyroid Hormones/genetics , Animals , Arginine/biosynthesis , Carrier Proteins/chemistry , Cell Line, Tumor , Cell Proliferation/genetics , Crystallography, X-Ray , Fibroblasts/metabolism , Glucose/metabolism , Glycolysis/genetics , Humans , Membrane Proteins/chemistry , Methylation , Mice , Mice, Knockout , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein-Arginine N-Methyltransferases/chemistry , Pyruvate Kinase/chemistry , Serine/genetics , Thyroid Hormones/chemistry , Thyroid Hormone-Binding Proteins
15.
Cell Rep ; 23(2): 389-403, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29641999

ABSTRACT

The arginine methylation status of histones dynamically changes during many cellular processes, including hematopoietic stem/progenitor cell (HSPC) development. The arginine methyltransferases and the readers that transduce the histone codes have been defined. However, whether arginine demethylation actively occurs in cells and what enzyme demethylates the methylarginine residues during various cellular processes are unknown. We report that JMJD1B, previously identified as a lysine demethylase for H3K9me2, mediates arginine demethylation of H4R3me2s and its intermediate, H4R3me1. We show that demethylation of H4R3me2s and H3K9me2s in promoter regions is correlated with active gene expression. Furthermore, knockout of JMJD1B blocks demethylation of H4R3me2s and/or H3K9me2 at distinct clusters of genes and impairs the activation of genes important for HSPC differentiation and development. Consequently, JMJD1B-/- mice show defects in hematopoiesis. Altogether, our study demonstrates that demethylase-mediated active arginine demethylation process exists in eukaryotes and that JMJD1B demethylates both H4R3me2s and H3K9me2 for epigenetic programming during hematopoiesis.


Subject(s)
Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Demethylation , Epigenesis, Genetic , Female , HEK293 Cells , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histones/chemistry , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/deficiency , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Substrate Specificity
16.
Nucleic Acids Res ; 46(6): 3061-3074, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29471495

ABSTRACT

DNA topoisomerase 3B (TOP3B) is unique among all mammalian topoisomerases for its dual activities that resolve both DNA and RNA topological entanglements to facilitate transcription and translation. However, the mechanism by which TOP3B activity is regulated is still elusive. Here, we have identified arginine methylation as an important post-translational modification (PTM) for TOP3B activity. Protein arginine methyltransferase (PRMT) 1, PRMT3 and PRMT6 all methylate TOP3B in vitro at its C-terminal arginine (R) and glycine (G)-rich motif. Site-directed mutagenesis analysis identified R833 and R835 as the major methylation sites. Using a methylation-specific antibody, we confirmed that TOP3B is methylated in cells and that mutation of R833 and R835 to lysine (K) significantly reduces TOP3B methylation. The methylation-deficient TOP3B (R833/835K) is less active in resolving negatively supercoiled DNA, which consequently lead to accumulation of co-transcriptionally formed R-loops in vitro and in cells. Additionally, the methylation-deficient TOP3B (R833/835K) shows reduced stress granule localization, indicating that methylation is critical for TOP3B function in translation regulation. Mechanistically, we found that R833/835 methylation is partially involved in the interaction of TOP3B with its auxiliary factor, the Tudor domain-containing protein 3 (TDRD3). Together, our findings provide the first evidence for the regulation of TOP3B activity by PTM.


Subject(s)
Amino Acid Motifs/genetics , Arginine/genetics , Cytoplasmic Granules/metabolism , DNA Topoisomerases, Type I/genetics , Protein Processing, Post-Translational , Amino Acid Sequence , Arginine/metabolism , Cell Line, Tumor , DNA Topoisomerases, Type I/metabolism , HeLa Cells , Humans , Methylation , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stress, Physiological
17.
Cell Discov ; 3: 16048, 2017.
Article in English | MEDLINE | ID: mdl-28101374

ABSTRACT

The Tudor domain-containing proteins are characterized by their specific interactions with methylated protein motifs, including methyl-arginines and methyl-lysines. The Tudor domain-containing protein 3 (TDRD3) is one of the major methyl-arginine effector molecules that recognizes methylated arginine residues on histones and the C-terminal domain of RNA polymerase II, and activates transcription. However, majority of the cellular TDRD3 localizes to the cytoplasm and its functions there are still elusive. Here, we have identified ubiquitin-specific protease 9 X-linked (USP9X) as a TDRD3-interacting protein by GST (glutathione S-transferase) pull-down and co-immunoprecipitation. Detailed characterization suggests that the interaction between TDRD3 and USP9X is mediated through the Tudor domain of TDRD3 and the arginine methylation of USP9X. This interaction plays a critical role in TDRD3 protein stability, as knockdown of USP9X expression leads to increased TDRD3 ubiquitination. We also found that USP9X co-localizes with TDRD3 in cytoplasmic stress granules and this localization is diminished in Tdrd3-null mouse embryonic fibroblast cells, suggesting that TDRD3 is essential for USP9X stress granule localization. Furthermore, we found that one of the USP9X de-ubiquitination targets, myeloid cell leukemia protein 1, is regulated by TDRD3, indicating that TDRD3 potentially regulates USP9X de-ubiquitinase activity. Finally, we show that knockdown of TDRD3 expression sensitizes breast cancer cells to chemotherapeutic drug-induced apoptosis, likely due to its regulation of USP9X. This study provides a novel candidate strategy for targeting apoptosis pathways in cancer therapy.

18.
Acta Biochim Biophys Sin (Shanghai) ; 48(7): 623-31, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27252122

ABSTRACT

The dynamic structure of chromatin, which exists in two conformational states: heterochromatin and euchromatin, alters the accessibility of the DNA to regulatory factors during transcription, replication, recombination, and DNA damage repair. Chemical modifications of histones and DNA, as well as adenosine triphospahate-dependent nucleosome remodeling, have been the major focus of research on chromatin dynamics over the past two decades. However, recent studies using a DNA-RNA hybrid-specific antibody and next-generation sequencing approaches have revealed that the formation of R-loops, one of the most common non-canonical DNA structures, is an emerging regulator of chromatin states. This review focuses on recent insights into the interplay between R-loop formation and the epigenetic modifications of chromatin in normal and disease states.


Subject(s)
Chromatin/metabolism , Chromatin/chemistry , DNA Damage , DNA Repair , DNA Replication , Protein Conformation , Recombination, Genetic , Transcription, Genetic
19.
Shanghai Kou Qiang Yi Xue ; 24(4): 447-50, 2015 Aug.
Article in Chinese | MEDLINE | ID: mdl-26383569

ABSTRACT

PURPOSE: To investigate the effect of three different zirconia angular abutments on the stress distribution in bone and abutment using three-dimensional finite element analysis, and provide instruction for clinical application. METHODS: Finite element analysis (FEA) was applied to analyze the stress distribution of three different zirconia/titanium angular abutments and bone around implant. RESULTS: The maximum Von Minses stress that existed in abutment, bolt and bone of the angular abutment model was significantly higher than that existed in the straight abutment model. The maximum Von Minses stress that existed in abutment, bolt and bone of the 20 ° angular abutment model was significantly higher than that existed in 15 ° angular abutment model. There was no significant difference between zirconia abutment model and titanium abutment model. CONCLUSIONS: The abutment angulation has a significant influence on the stress distribution in the abutment, bolt and bone, and exacerbates as the angulation increases, which suggest that we should take more attention to the implant orientation and use straight abutment or little angular abutment. The zirconia abutment can be used safely, and there is no noticeable difference between zirconia abutment and titanium abutment on stress distribution.


Subject(s)
Dental Abutments , Finite Element Analysis , Zirconium/chemistry , Alveolar Process , Dental Stress Analysis , Humans , Maxilla , Stress, Mechanical , Titanium
20.
Shanghai Kou Qiang Yi Xue ; 24(3): 370-2, 2015 Jun.
Article in Chinese | MEDLINE | ID: mdl-26166532

ABSTRACT

PURPOSE: To evaluate the effect of N2O inhalation and oral midazolam sedation on uncooperative patients with intellectual disability in pediatric dentistry. METHODS: N2O inhalation (35%-50%) and oral midazolam conscious sedation (dosages range: 0.50-0.75 mg/kg) were applied to 67 uncooperative pediatric patients with intellectual disability in outpatient department. The patients were divided into 2 groups: group A (N2O inhalation conscious sedation) and group B(oral midazolam conscious sedation).Treatment results and safety were statistically analyzed by Chi-square test with SPSSl3.0 software package. RESULTS: The mean success rate was 70%. The success rate in group B (75%) was higher than group A (67%). The overall incidence of adverse reactions was 13%(9/67). The adverse reaction rate in group B (25%) was significantly higher than group A (5%, P<0.05). CONCLUSIONS: N2O inhalation and oral midazolam conscious sedation are effective and safe in pediatric dental uncooperative patients with intellectual disability.


Subject(s)
Conscious Sedation , Intellectual Disability , Administration, Inhalation , Child , Humans , Midazolam , Nitrogen Oxides , Pediatric Dentistry
SELECTION OF CITATIONS
SEARCH DETAIL
...