Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
J Pharmacol Sci ; 154(3): 209-217, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38395522

ABSTRACT

Upregulation of nitric oxide (NO) production contributes to the pathogenesis of numerous diseases via S-nitrosylation, a post-translational modification of proteins. This process occurs due to the oxidative reaction between NO and a cysteine thiol group; however, the extent of this reaction remains unknown. S-Nitrosylation of PRMT1, a major asymmetric arginine methyltransferase of histones and numerous RNA metabolic proteins, was induced by NO donor treatment. We found that nitrosative stress leads to S-nitrosylation of cysteine 119, located near the active site, and attenuates the enzymatic activity of PRMT1. Interestingly, RNA sequencing analysis revealed similarities in the changes in expression elicited by NO and PRMT1 inhibitors or knockdown. A comprehensive search for PRMT1 substrates using the proximity-dependent biotin identification method highlighted many known and new substrates, including RNA-metabolizing enzymes. To validate this result, we selected the RNA helicase DDX3 and demonstrated that arginine methylation of DDX3 is induced by PRMT1 and attenuated by NO treatment. Our results suggest the existence of a novel regulatory system associated with transcription and RNA metabolism via protein S-nitrosylation.


Subject(s)
Arginine , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Arginine/metabolism , Cysteine , Histones/metabolism , RNA
2.
J Biol Chem ; 296: 100417, 2021.
Article in English | MEDLINE | ID: mdl-33587951

ABSTRACT

Induction of fetal hemoglobin to overcome adult ß-globin gene deficiency is an effective therapeutic strategy to ameliorate human ß-hemoglobinopathies. Previous work has revealed that fetal γ-globin can be translationally induced via integrated stress signaling, but other studies have indicated that activating stress may eventually suppress γ-globin expression transcriptionally. The mechanism by which γ-globin expression is regulated at the translational level remains largely unknown, limiting our ability to determine whether activating stress is a realistic therapeutic option for these disorders. In this study, we performed a functional CRISPR screen targeting protein arginine methyltransferases (PRMTs) to look for changes in γ-globin expression in K562 cells. We not only discovered that several specific PRMTs may block γ-globin transcription, but also revealed PRMT1 as a unique family member that is able to suppress γ-globin synthesis specifically at the translational level. We further identified that a non-AUG uORF within the 5' untranslated region of γ-globin serves as a barrier for translation, which is bypassed upon PRMT1 deficiency. Finally, we found that this novel mechanism of γ-globin suppression could be pharmacologically targeted by the PRMT1 inhibitor, furamidine dihydrochloride. These data raise new questions regarding methyltransferase function and may offer a new therapeutic direction for ß-hemoglobinopathies.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , gamma-Globins/metabolism , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Fetal Hemoglobin/pharmacology , Gene Expression/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , K562 Cells , Methyltransferases/metabolism , Protein Biosynthesis/genetics , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , beta-Globins/metabolism , gamma-Globins/genetics
3.
Biochim Biophys Acta Gen Subj ; 1865(1): 129776, 2021 01.
Article in English | MEDLINE | ID: mdl-33127433

ABSTRACT

BACKGROUND: Protein arginine methyltransferase 1 (PRMT1), a major type I arginine methyltransferase in mammals, methylates histone and non-histone proteins to regulate various cellular functions such as transcription, DNA damage response, and signal transduction. SCOPE OF REVIEW: This review summarizes previous and recent studies on PRMT1 functions in major cell types of the central nervous system. We also discuss the potential involvement of PRMT1 in neurodegenerative diseases such as amyotrophic lateral sclerosis and frontotemporal dementia. Also, we raise key questions that must be addressed in the future to more precisely understand the roles of PRMT1. MAJOR CONCLUSIONS: Recent studies revealed that PRMT1 is essential for the development of neurons, astrocytes, and oligodendrocytes, although further investigation using cell type-specific PRMT1-deficient animals is required. In addition, the relevance of PRMT1 in neurodegenerative diseases will continue to be a hot topic. GENERAL SIGNIFICANCE: PRMT1 is important for neural development and neurodegenerative diseases.


Subject(s)
Arginine/metabolism , Brain/growth & development , Neurodegenerative Diseases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Animals , Arginine/analogs & derivatives , Arginine/genetics , Brain/metabolism , Brain/pathology , Gene Expression Regulation, Developmental , Humans , Methylation , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics
4.
DNA Repair (Amst) ; 95: 102953, 2020 11.
Article in English | MEDLINE | ID: mdl-32861926

ABSTRACT

The up-regulation of PRMT1 is critical to the cell growth and cancer progression of lung cancer cells. In our research, we found that PRMT1 is important to the DNA repair ability and drug resistance of lung cancer cells. To demonstrate the functions of PRMT1, we identified Flap endonuclease 1 (FEN1) as a post-translationally modified downstream target protein of PRMT1. As a major component of Base Excision Repair pathway, FEN1 plays an important role in DNA replication and DNA damage repair. However, the detailed mechanism of FEN1 up-regulation in lung cancer cells remains unclear. In our study, we identified PRMT1 as a key factor that maintains the high expression levels of FEN1, which is critical to the DNA repair ability and the chemotherapeutic drug resistance of lung cancer cells.


Subject(s)
Drug Resistance, Neoplasm/genetics , Flap Endonucleases/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , A549 Cells , Apoptosis/genetics , Cell Proliferation/genetics , DNA Repair , Epigenesis, Genetic , Gene Knockdown Techniques , Humans , Protein-Arginine N-Methyltransferases/deficiency , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/deficiency , Repressor Proteins/genetics , Up-Regulation
5.
Int J Mol Sci ; 21(9)2020 Apr 26.
Article in English | MEDLINE | ID: mdl-32357521

ABSTRACT

Protein arginine methyltransferase 1 (PRMT1) is the most predominant PRMT and is type I, meaning it generates monomethylarginine and asymmetric dimethylarginine. PRMT1 has functions in oxidative stress, inflammation and cancers, and modulates diverse diseases; consequently, numerous trials to develop PRMT1 inhibitors have been attempted. One selective PRMT1 inhibitor is N,N'-(Sulfonyldi-4,1-phenylene)bis(2-chloroacetamide), also named TC-E 5003 (TC-E). In this study, we investigated whether TC-E regulated inflammatory responses. Nitric oxide (NO) production was evaluated by the Griess assay and the inflammatory gene expression was determined by conducting RT-PCR. Western blot analyzing was carried out for inflammatory signaling exploration. TC-E dramatically reduced lipopolysaccharide (LPS)-induced NO production and the expression of inflammatory genes (inducible NO synthase (iNOS), cyclooxygenase (COX)-2, tumor necrosis factor (TNF)-α and interleukin (IL)-6) as determined using RT-PCR. TC-E downregulated the nuclear translocation of the nuclear factor (NF)-κB subunits p65 and p50 and the activator protein (AP)-1 transcriptional factor c-Jun. Additionally, TC-E directly regulated c-Jun gene expression following LPS treatment. In NF-κB signaling, the activation of IκBα and Src was attenuated by TC-E. Taken together, these data show that TC-E modulates the lipopolysaccharide (LPS)-induced AP-1 and NF-κB signaling pathways and could possibly be further developed as an anti-inflammatory compound.


Subject(s)
Acetamides/pharmacology , Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides/adverse effects , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Acetamides/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Molecular Structure , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Front Oncol ; 10: 226, 2020.
Article in English | MEDLINE | ID: mdl-32231994

ABSTRACT

About 30% of medulloblastomas (MBs), a tumor of the cerebellum, arise from cerebellar granule cell precursors (GCPs) undergoing transformation following activation of the Sonic hedgehog (Shh) pathway. To study this process, we generated a new MB model by crossing Patched1 heterozygous (Ptch1 +/-) mice, which develop spontaneous Shh-type MBs, with mice lacking B-cell translocation gene 1 (Btg1), a regulator of cerebellar development. In MBs developing in Ptch1 +/- mice, deletion of Btg1 does not alter tumor and lesion frequencies, nor affect the proliferation of neoplastic precursor cells. However, in both tumors and lesions arising in Ptch1 +/- mice, ablation of Btg1 increases by about 25% the apoptotic neoplastic precursor cells, as judged by positivity to activated caspase-3. Moreover, although Btg1 ablation in early postnatal GCPs, developing in the external granule cell layer, leads to a significant increase of proliferation, and decrease of differentiation, relative to wild-type, no synergy occurs with the Ptch1 +/- mutation. However, Btg1 deletion greatly increases apoptosis in postnatal GCPs, with strong synergy between Btg1-null and Ptch1 +/- mutations. That pronounced increase of apoptosis observed in Ptch1 +/- /Btg1 knockout young or neoplastic GCPs may be responsible for the lack of effect of Btg1 ablation on tumorigenesis. This increased apoptosis may be a consequence of increased expression of protein arginine methyltransferase 1 (Prmt1) protein that we observe in Btg1 knockout/Ptch1 +/- MBs. In fact, apoptotic genes, such as BAD, are targets of Prmt1. Moreover, in Btg1-null MBs, we observed a two-fold increase of cells positive to CD15, which labels tumor stem cells, raising the possibility of activation of quiescent tumor cells, known for their role in long-term resistance to treatment and relapses. Thus, Btg1 appears to play a role in cerebellar tumorigenesis by regulating the balance between apoptosis and proliferation during MB development, also influencing the number of tumor stem cells.

7.
J Biol Chem ; 295(20): 7126-7137, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32245889

ABSTRACT

Protein arginine methyltransferase 1 (PRMT1) is a key regulator of hepatic immune responses. Recently, we reported that PRMT1 regulates the tumor immune response in hepatocellular carcinoma (HCC). Here we found that PRMT1 expression in human HCC correlates with that of programmed cell death 1 ligand 1 (PD-L1), PD-L2, and other checkpoint genes. PRMT1 deletion in mice reduced PD-L1 and PD-L2 expression in tumors and reduced the efficiency of PD-1 antibody treatment in a diethylnitrosamine-induced HCC mouse model, suggesting that PRMT1 regulates the hepatic immune checkpoint. Mice had reduced PD-L1 and PD-L2 expression when PRMT1 was specifically deleted in tumor cells or macrophages, but PRMT1 deletion in dendritic cells did not alter PD-L1 and PD-L2 expression. rs975484 is a common polymorphism in the human PRMT1 gene promoter, and we found that it alters PRMT1 expression in blood monocytes and tumor-associated macrophages in human HCC. PRMT1 expression was higher in individuals with a GG genotype than in individuals with a CC genotype, and heterozygous carriers had intermediate expression. Luciferase reporter assays indicated that this differential expression is due to an extra C/EBPß-binding site in the PRMT1 promoter of individuals carrying the minor G allele. The rs975484 genotype also correlated with PRMT1 target expression in HCC. Individuals with the GG genotype had significantly higher levels of the PRMT1 targets PD-L1, PD-L2, and VISTA than those with the CC genotype. We conclude that PRMT1 critically controls immune checkpoints in mice and humans and that the PRMT1 polymorphism rs975484 affects checkpoint gene expression in HCC.


Subject(s)
B7 Antigens/immunology , B7-H1 Antigen/immunology , Carcinoma, Hepatocellular/immunology , Gene Expression Regulation, Neoplastic/immunology , Liver Neoplasms/immunology , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Protein-Arginine N-Methyltransferases/immunology , Repressor Proteins/immunology , Animals , B7 Antigens/genetics , B7-H1 Antigen/genetics , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Diethylnitrosamine/toxicity , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Programmed Cell Death 1 Ligand 2 Protein/genetics , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , THP-1 Cells
8.
Biochim Biophys Acta Gen Subj ; 1864(3): 129509, 2020 03.
Article in English | MEDLINE | ID: mdl-31884067

ABSTRACT

BACKGROUND: Brains express structurally unique glycans, including human natural killer-1 (HNK-1), which participate in development and high-order functions. However, the regulatory mechanisms of expression of these brain-specific glycans are largely unknown. We examined whether arginine methylation, another type of protein modification essential for neural development, impacts the expression of various glycans in the developing brain. METHODS: We analyzed several types of glycans, including the HNK-1 epitope, in the cerebellum and cerebral cortex from mice with nervous system-specific knockout of protein arginine methyltransferase 1 (PRMT1). We also analyzed the expression levels of glycosyltransferases responsible for HNK-1 and of HNK-1 carrier glycoproteins by quantitative RT-PCR and western blotting. RESULTS: Among several glycans, expression of HNK-1 glycan was strikingly upregulated in the PRMT1-deficient cerebellum. Furthermore, such upregulation was found in the cerebellum but not in the cerebral cortex. Regarding the mechanisms, we demonstrated that the mRNA level and activity of the responsible glycosyltransferase (B3gat1) were elevated in the knockout cerebellum. We also showed that the expression of HNK-1 carrier glycoproteins such as neural cell adhesion molecule (NCAM), L1 and AMPA receptor subunit GluA2 were also increased in the PRMT1-deficient cerebellum. CONCLUSIONS: Loss of arginine methylation leads to an increase in HNK-1 glycan in the developing cerebellum but not in the cerebral cortex via upregulation of the biosynthetic enzyme and carrier glycoproteins. GENERAL SIGNIFICANCE: PRMT1 is a novel regulator of HNK-1 glycan production in the cerebellum. Mechanisms involving crosstalk between glycosylation and arginine methylation are suggested to occur.


Subject(s)
CD57 Antigens/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Animals , Brain , Cerebellum , Cerebral Cortex , Epitopes/genetics , Female , Glucuronosyltransferase/metabolism , Glycosylation , Glycosyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polysaccharides/metabolism , Protein-Arginine N-Methyltransferases/genetics , Receptors, AMPA/genetics , Repressor Proteins/genetics , Transcriptional Activation
9.
Placenta ; 65: 47-54, 2018 05.
Article in English | MEDLINE | ID: mdl-29908641

ABSTRACT

INTRODUCTION: In mammals, the placenta is an organ that is required to maintain the development of fetus during pregnancy. Although the proper formation of placenta is in part regulated by the post-translational modifications of proteins, little is known regarding protein arginine methylation during placental development. Here, we characterized developmental expression of protein arginine methyltransferase 1 (PRMT1) in mouse placentas. METHODS: Expression levels of PRMT1 mRNA and protein in placentas were investigated using the real-time quantitative PCR and Western blot, respectively. Next, the localization of PRMT1 was determined by immunohistochemistry and immunofluorescence analyses. In addition, the levels of methylarginines of placental proteins were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS: PRMT1 mRNA and its protein were expressed at highest levels in mid-gestation stages, and their expression showed stepwise decrease in the late gestation. At embryonic (E) day 9, PRMT1 was observed in several different trophoblast cell (TC) subtypes. Furthermore, PRMT1 was mainly expressed in the labyrinth zone of TCs at E13. Finally, total methylarginines of proteins were significantly reduced in late gestation of placentas compared with mid-gestation stages. DISCUSSION: In this study, we found developmental changes in the placental expression of PRMT1 and in protein arginine methylation status during pregnancy. These findings provide fundamental information regarding placental PRMT1-mediated arginine methylation during the development.


Subject(s)
Placenta/metabolism , Placentation/genetics , Protein-Arginine N-Methyltransferases/genetics , Animals , Arginine/metabolism , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Placenta/enzymology , Pregnancy , Protein Processing, Post-Translational/genetics , Protein-Arginine N-Methyltransferases/metabolism
10.
Acta Neuropathol ; 131(4): 587-604, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26895297

ABSTRACT

Deposition of the nuclear DNA/RNA-binding protein Fused in sarcoma (FUS) in cytosolic inclusions is a common hallmark of some cases of frontotemporal lobar degeneration (FTLD-FUS) and amyotrophic lateral sclerosis (ALS-FUS). Whether both diseases also share common pathological mechanisms is currently unclear. Based on our previous finding that FUS deposits are hypomethylated in FTLD-FUS but not in ALS-FUS, we have now investigated whether genetic or pharmacological inactivation of Protein arginine methyltransferase 1 (PRMT1) activity results in unmethylated FUS or in alternatively methylated forms of FUS. To do so, we generated FUS-specific monoclonal antibodies that specifically recognize unmethylated arginine (UMA), monomethylated arginine (MMA) or asymmetrically dimethylated arginine (ADMA). Loss of PRMT1 indeed not only results in an increase of UMA FUS and a decrease of ADMA FUS, but also in a significant increase of MMA FUS. Compared to ADMA FUS, UMA and MMA FUS exhibit much higher binding affinities to Transportin-1, the nuclear import receptor of FUS, as measured by pull-down assays and isothermal titration calorimetry. Moreover, we show that MMA FUS occurs exclusively in FTLD-FUS, but not in ALS-FUS. Our findings therefore provide additional evidence that FTLD-FUS and ALS-FUS are caused by distinct disease mechanisms although both share FUS deposits as a common denominator.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Lobar Degeneration/metabolism , Protein-Arginine N-Methyltransferases/metabolism , RNA-Binding Protein FUS/metabolism , beta Karyopherins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Antibodies/pharmacology , Arginine/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Embryonic Stem Cells , Enzyme Inhibitors/pharmacology , Female , Frontotemporal Lobar Degeneration/genetics , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Protein Binding/drug effects , Protein Binding/genetics , Protein-Arginine N-Methyltransferases/genetics , RNA-Binding Protein FUS/immunology , Rats , beta Karyopherins/immunology
11.
J Proteome Res ; 14(11): 4571-80, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26398198

ABSTRACT

Liver receptor homologue-1 (LRH1) is an orphan nuclear receptor that has been shown to play a role in the transcriptional regulation of pathways involved in cancer. Elucidating the components of the LRH1 transcriptional complex to better understand endogenous regulation of the receptor as well as its role in cancer remains a high priority. A sub-cellular enrichment strategy coupled with proteomic approaches was employed to identify putative LRH1 co-regulators. Nuclear fractionation protocol was essential for detection of LRH1 peptides by mass spectrometry (MS), with most peptides being observed in the insoluble fraction (receptor bound to DNA). SERBP1 and ILF3 were identified as LRH1 interacting partners by both Western blot and MS/MS analysis. Receptor knockdown by siRNA showed an increase in SERBP1 expression, while ILF3 expression was unchanged. In contrast, receptor overexpression decreased only SERBP1 mRNA levels. Consistent with these data, in a promoter:reporter assay, binding of LRH1 to the promoter region of SERBP1 resulted in a decrease in the expression level of the reporter gene, subsequently inhibiting transcription. Given the receptor's role in cancer progression, the study here elucidates additional transcriptional machinery involved in LRH1 signaling and potentially provides new targets for therapeutics development.


Subject(s)
Gene Expression Regulation , Peptides/analysis , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic , Amino Acid Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Chemical Fractionation , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Molecular Sequence Annotation , Molecular Sequence Data , Nuclear Factor 90 Proteins/genetics , Nuclear Factor 90 Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-595179

ABSTRACT

Objective:To investigate the arginine (Arg) sites in splicing factor 2/alternative splicing factor (SF2/ASF) methylated by protein arginine methyltransferase 1 (PRMT1). Methods:Wild-type and Arg93,Arg97,Arg109 mutant SF2/ASF plasmids were constructed,and GST-PRMT1,GST-SF2/ASF and arginine mutant GST-SF2/ASF fusion proteins were induced and purified. Methylation activity of PRMT1 on wild-type or mutant SF2/ASF protein and methylated sites of SF2/ASF were examined by methylation assay. The effect of SF2/ASF methylation on its subcellular localization was analyzed by immunofluorescence assay.Results:PRMT1 induced methylation of SF2/ASF at arginine,and PRMT1 did not methylate SF2/ASF when SF2/ASF was mutant at Arg93,Arg97 or Arg109,with Arg97 mutation showing the most profound inhibitory effect. Methylation of SF2/ASF did not affect its subcellular localization.Conclusion:SF2/ASF is a newly identified substrate of PRMT1; Arg93,Arg97 and Arg 109 are the three methylation sites in SF2/ASF,and Arg97 is the main methylation site. Methylation of SF2/ASF does not affect its subcellular localization.

SELECTION OF CITATIONS
SEARCH DETAIL