Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 127
1.
Mol Cell ; 81(17): 3481-3495.e7, 2021 09 02.
Article En | MEDLINE | ID: mdl-34358446

PRMT5 is an essential arginine methyltransferase and a therapeutic target in MTAP-null cancers. PRMT5 uses adaptor proteins for substrate recruitment through a previously undefined mechanism. Here, we identify an evolutionarily conserved peptide sequence shared among the three known substrate adaptors (CLNS1A, RIOK1, and COPR5) and show that it is necessary and sufficient for interaction with PRMT5. We demonstrate that PRMT5 uses modular adaptor proteins containing a common binding motif for substrate recruitment, comparable with other enzyme classes such as kinases and E3 ligases. We structurally resolve the interface with PRMT5 and show via genetic perturbation that it is required for methylation of adaptor-recruited substrates including the spliceosome, histones, and ribosomal complexes. Furthermore, disruption of this site affects Sm spliceosome activity, leading to intron retention. Genetic disruption of the PRMT5-substrate adaptor interface impairs growth of MTAP-null tumor cells and is thus a site for development of therapeutic inhibitors of PRMT5.


Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/physiology , Animals , Cell Line, Tumor , Cytoplasm/metabolism , Female , HCT116 Cells , HEK293 Cells , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ion Channels/metabolism , Male , Methylation , Mice , Mice, Nude , Nuclear Proteins/metabolism , Peptides/genetics , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Spliceosomes/metabolism
2.
Elife ; 102021 08 27.
Article En | MEDLINE | ID: mdl-34448450

Protein arginine methyltransferase 5 (Prmt5) is the major type II enzyme responsible for symmetric dimethylation of arginine. Here, we found that PRMT5 was expressed at high level in ovarian granulosa cells of growing follicles. Inactivation of Prmt5 in granulosa cells resulted in aberrant follicle development and female infertility. In Prmt5-knockout mice, follicle development was arrested with disorganized granulosa cells in which WT1 expression was dramatically reduced and the expression of steroidogenesis-related genes was significantly increased. The premature differentiated granulosa cells were detached from oocytes and follicle structure was disrupted. Mechanism studies revealed that Wt1 expression was regulated by PRMT5 at the protein level. PRMT5 facilitated IRES-dependent translation of Wt1 mRNA by methylating HnRNPA1. Moreover, the upregulation of steroidogenic genes in Prmt5-deficient granulosa cells was repressed by Wt1 overexpression. These results demonstrate that PRMT5 participates in granulosa cell lineage maintenance by inducing Wt1 expression. Our study uncovers a new role of post-translational arginine methylation in granulosa cell differentiation and follicle development.


Infertility in women can be caused by many factors, such as defects in the ovaries. An important part of the ovaries for fertility are internal structures called follicles, which house early forms of egg cells. A follicle grows and develops until the egg is finally released from the ovary into the fallopian tube, where the egg can then be fertilised. In the follicle, an egg is surrounded by other types of cells, such as granulosa cells. The egg and neighbouring cells must maintain healthy contacts with each other, otherwise the follicle can stop growing and developing, potentially causing infertility. The development of a follicle depends on an array of proteins. For example, the transcription factor WT1 controls protein levels by activating other genes and their proteins and is produced in high numbers by granulosa cells at the beginning of follicle development. Although WT1 levels dip towards the later stages of follicle development, insufficient levels can lead to defects. So far, it has been unclear how levels of WT1in granulose cells are regulated. Chen, Dong et al. studied mouse follicles to reveal more about the role of WT1 in follicle development. The researchers measured protein levels in mouse granulosa cells as the follicles developed, and discovered elevated levels of PRMT5, a protein needed for egg cells to form and survive in the follicles. Blocking granulosa cells from producing PRMT5 led to abnormal follicles and infertility in mice. Moreover, mice that had been engineered to lack PRMT5 developed abnormal follicles, where the egg and surrounding granulosa cells were not attached to each other, and the granulosa cells had low levels of WT1. Further experiments revealed that PRMT5 controlled WT1 levels by adding small molecules called methyl groups to another regulatory protein called HnRNPA1. The addition of methyl groups to genes or their proteins is an important modification that takes place in many processes within a cell. Chen, Dong et al. reveal that this activity also plays a key role in maintaining healthy follicle development in mice, and that PRMT5 is necessary for controlling WT1. Identifying all of the intricate mechanism involved in regulating follicle development is important for finding ways to combat infertility.


Ovarian Follicle/growth & development , Protein-Arginine N-Methyltransferases/physiology , WT1 Proteins/genetics , Animals , Female , Infertility, Female/genetics , Mice , Mice, Knockout , Mice, Transgenic , Protein-Arginine N-Methyltransferases/genetics
3.
Gene ; 791: 145718, 2021 Jul 30.
Article En | MEDLINE | ID: mdl-33991650

The incidence rates of colorectal cancer have been increasing in the last decades, yet the overall survival rate is still not ideal. There is a need to further investigate detailed mechanism for colorectal cancer tumorigenesis. The biological function of protein arginine methyltransferases 3 (PRMT3) is seldom studied in tumorigenesis. Here, we attempted to elucidate the link between PRMT3 and tumorigenesis in colorectal cancer. Results revealed that PRMT3 was upregulated in colorectal cancer. Besides, PRMT3 overexpression promoted colorectal cancer cell proliferation, migration, and invasion. Regarding mechanism for colorectal cancer tumorigenesis, PRMT3 stabilized C-MYC and the pro-tumorigenesis function of PRMT3 was dependent on C-MYC. Clinically, these findings might provide a novel therapeutical treatment strategy for colorectal cancer.


Colorectal Neoplasms/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Methylation , Neoplasm Invasiveness/genetics , Protein-Arginine N-Methyltransferases/physiology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Ribosomal Proteins/metabolism
4.
Theranostics ; 11(13): 6592-6606, 2021.
Article En | MEDLINE | ID: mdl-33995678

Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.


Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Nuclear Proteins/deficiency , Receptor, Transforming Growth Factor-beta Type II/antagonists & inhibitors , Trans-Activators/deficiency , Adaptor Proteins, Signal Transducing/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Code , Humans , Lung Neoplasms/genetics , Methylation , Mice, Transgenic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplastic Stem Cells/pathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/physiology , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/physiology , Tumor Burden
5.
Endocrinology ; 162(6)2021 06 01.
Article En | MEDLINE | ID: mdl-33713122

Melatonin, a pineal gland hormone, has been suggested to treat postmenopausal osteoporosis due to its inhibitory effect on osteoclast differentiation. We previously reported that protein arginine methyltransferase 1 (PRMT1) was an important mediator of receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis. However, the relationship between melatonin and PRMT1 in osteoclast differentiation and estrogen deficiency-induced osteoporosis is unclear. In this study, we investigated the inhibitory mechanisms of melatonin in vitro and in vivo by focusing on PRMT1. Melatonin treatment effectively blocked RANKL-induced osteoclastogenesis by inhibiting PRMT1 and asymmetric dimethylarginine (ADMA) expression. RANKL-induced tumor necrosis factor receptor-associated factor 6 (TRAF6) and the phosphorylation of JNK were also suppressed by melatonin, and TRAF6 siRNA attenuated RANKL-induced p-JNK and PRMT1 production. Melatonin inhibited the transcriptional activity of NF-κB by interfering with the binding of PRMT1 and NF-κB subunit p65 in RANKL-treated bone marrow-derived macrophages. Our results also revealed that melatonin inhibits RANKL-induced PRMT1 expression through receptors-independent pathway. Thus, the anti-osteoclastogenic effect of melatonin was mediated by a cascade of inhibition of RANKL-induced TRAF6, JNK, PRMT1, and NF-κB signaling in melatonin receptors-independent pathway. In vivo, ovariectomy caused significant decreases in bone mineral density, but melatonin treatment alleviated the ovariectomized (OVX)-induced bone loss by inhibiting bone resorption. Furthermore, the expression PRMT1 and TRAP mRNA was upregulated in OVX-femurs, but effectively suppressed by melatonin injection. These findings suggest that melatonin inhibited osteoclast differentiation and estrogen deficiency-induced osteoporosis by suppressing RANKL-induced TRAF6, JNK, PRMT1, and NF-κB signaling cascades in melatonin receptors-independent pathway.


Bone Resorption/prevention & control , Melatonin/pharmacology , Osteoclasts/drug effects , Animals , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Bone Diseases, Metabolic/prevention & control , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , Osteoclasts/physiology , Osteogenesis/drug effects , Ovariectomy/adverse effects , Protein-Arginine N-Methyltransferases/physiology , Signal Transduction/drug effects , Signal Transduction/genetics
6.
J Biol Chem ; 296: 100417, 2021.
Article En | MEDLINE | ID: mdl-33587951

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.


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
7.
Commun Biol ; 4(1): 109, 2021 01 25.
Article En | MEDLINE | ID: mdl-33495566

Protein arginine methyltransferase 3 (PRMT3) regulates protein functions by introducing asymmetric dimethylation marks at the arginine residues in proteins. However, very little is known about the interaction partners of PRMT3 and their functional outcomes. Using yeast-two hybrid screening, we identified Retinal dehydrogenase 1 (ALDH1A1) as a potential interaction partner of PRMT3 and confirmed this interaction using different methods. ALDH1A1 regulates variety of cellular processes by catalyzing the conversion of retinaldehyde to retinoic acid. By molecular docking and site-directed mutagenesis, we identified the specific residues in the catalytic domain of PRMT3 that facilitate interaction with the C-terminal region of ALDH1A1. PRMT3 inhibits the enzymatic activity of ALDH1A1 and negatively regulates the expression of retinoic acid responsive genes in a methyltransferase activity independent manner. Our findings show that in addition to regulating protein functions by introducing methylation modifications, PRMT3 could also regulate global gene expression through protein-protein interactions.


Aldehyde Dehydrogenase 1 Family/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Retinal Dehydrogenase/metabolism , Tretinoin/metabolism , Down-Regulation/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Protein Binding , Protein-Arginine N-Methyltransferases/physiology , Signal Transduction/drug effects , Tretinoin/pharmacology
8.
Cell Rep ; 33(10): 108490, 2020 12 08.
Article En | MEDLINE | ID: mdl-33296647

Cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS), upon sensing cytosolic DNA, catalyzes the production of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which activates STING-TBK1-IRF3 signaling. cGAS is also present in the nucleus, but the relevant nuclear function or mechanism remains largely unknown. Here, we report that nuclear cGAS is indispensable for inducing cytokines and chemokines triggered by RNA/DNA viruses. Unexpectedly, the DNA-binding/nucleotidyltransferase activity of cGAS is dispensable for RNA-virus-induced genes expression. cGAS deficiency does not affect the phosphorylation, dimerization, or nuclear translocation of IRF3 induced by double-stranded RNA (dsRNA). Mechanistically, nuclear-localized cGAS interacts with protein arginine methyltransferase 5 (Prmt5), which catalyzes the symmetric dimethylation of histone H3 arginine 2 at Ifnb and Ifna4 promoters, thus facilitating the access of IRF3. Deficiency of Prmt5 or disrupting its catalytic activity suppresses the production of type I interferons (IFNs), impairing the host defenses against RNA/DNA virus infections. Taken together, our study uncovers a non-canonical function of nuclear-localized cGAS in innate immunity via regulating histone arginine modification.


Nucleotidyltransferases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Antiviral Agents/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclic GMP/metabolism , Cytosol/metabolism , Female , Humans , Immunity, Innate/genetics , Interferon Type I/metabolism , Interferon-beta/metabolism , Male , Membrane Proteins/metabolism , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotides, Cyclic , Nucleotidyltransferases/physiology , Phosphorylation/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/physiology , Signal Transduction/physiology
9.
Cancer Res ; 80(22): 4904-4917, 2020 11 15.
Article En | MEDLINE | ID: mdl-32999000

The majority of advanced prostate cancer therapies aim to inhibit androgen receptor (AR) signaling. However, AR reactivation inevitably drives disease progression to castration-resistant prostate cancer (CRPC). Here we demonstrate that protein arginine methyltransferase 5 (PRMT5) functions as an epigenetic activator of AR transcription in CRPC, requiring cooperation with a methylosome subunit pICln. In vitro and in xenograft tumors in mice, targeting PRMT5 or pICln suppressed growth of CRPC cells. Full-length AR and AR-V7 transcription activation required both PRMT5 and pICln but not MEP50. This activation of transcription was accompanied by PRMT5-mediated symmetric dimethylation of H4R3 at the proximal AR promoter. Further, knockdown of PRMT5 abolished the binding of pICln (but not vice versa) to the AR proximal promoter region, suggesting that PRMT5 recruits pICln to the AR promoter to activate AR transcription. Differential gene expression analysis in 22Rv1 cells confirmed that PRMT5 and pICln both regulate the androgen signaling pathway. In addition, PRMT5 and pICln protein expression positively correlated with AR and AR-V7 protein expression in CRPC tissues and their expression was highly correlated at the mRNA level across multiple publicly available CRPC datasets. Our results suggest that targeting PRMT5 or pICln may be explored as a novel therapy for CRPC treatment by suppressing expression of AR and AR splice variants to circumvent AR reactivation. SIGNIFICANCE: This study provides evidence that targeting PRMT5 can eliminate expression of AR and can be explored as a novel therapeutic approach to treat metastatic hormone-naïve and castration-resistant prostate cancer.


Ion Channels/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Protein-Arginine N-Methyltransferases/physiology , Receptors, Androgen/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Enlargement , Disease Progression , Down-Regulation , Epigenesis, Genetic/physiology , Gene Expression Profiling , Gene Knockdown Techniques , Heterografts , Humans , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Male , Methylation , Mice , Neoplasm Transplantation , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Receptors, Histamine H2/metabolism , Receptors, Histamine H3/metabolism
10.
Mol Biol Cell ; 31(18): 1963-1973, 2020 08 15.
Article En | MEDLINE | ID: mdl-32583741

Arginine methylation is a common posttranslational modification that modulates protein function. SCY1-like pseudokinase 1 (SCYL1) is crucial for neuronal functions and interacts with γ2-COP to form coat protein complex I (COPI) vesicles that regulate Golgi morphology. However, the molecular mechanism by which SCYL1 is regulated remains unclear. Here, we report that the γ2-COP-binding site of SCYL1 is arginine-methylated by protein arginine methyltransferase 1 (PRMT1) and that SCYL1 arginine methylation is important for the interaction of SCYL1 with γ2-COP. PRMT1 was colocalized with SCYL1 in the Golgi fraction. Inhibition of PRMT1 suppressed axon outgrowth and dendrite complexity via abnormal Golgi morphology. Knockdown of SCYL1 by small interfering RNA (siRNA) inhibited axon outgrowth, and the inhibitory effect was rescued by siRNA-resistant SCYL1, but not SCYL1 mutant, in which the arginine methylation site was replaced. Thus, PRMT1 regulates Golgi morphogenesis via SCYL1 arginine methylation. We propose that SCYL1 arginine methylation by PRMT1 contributes to axon and dendrite morphogenesis in neurons.


Adaptor Proteins, Vesicular Transport/metabolism , Coatomer Protein/metabolism , DNA-Binding Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Vesicular Transport/physiology , Animals , Arginine/metabolism , Coat Protein Complex I/metabolism , Coatomer Protein/physiology , DNA-Binding Proteins/physiology , Female , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Male , Methylation , Mice , Mice, Inbred ICR , Neuronal Outgrowth/physiology , Protein Processing, Post-Translational/drug effects , Protein-Arginine N-Methyltransferases/physiology , Rats , Rats, Wistar , Repressor Proteins/physiology , Transcription Factors/metabolism
11.
Theranostics ; 10(1): 133-150, 2020.
Article En | MEDLINE | ID: mdl-31903111

Hair cells in the inner ear have been shown to be susceptible to ototoxicity from some beneficial pharmaceutical drugs, such as aminoglycosides and cisplatin. Thus, there is great interest in discovering new targets or compounds that protect hair cells from these ototoxic drugs. Epigenetic regulation is closely related to inner ear development; however, little is known about epigenetic regulation in the process of ototoxic drugs-induced hearing loss. Methods: In this study, we investigated the role of protein arginine methyltransferase 6 (PRMT6) in aminoglycoside- and cisplatin-induced hair cell loss by using EPZ020411, a selective small molecule PRMT6 inhibitor, in vitro in neonatal mouse cochlear explants and in vivo in C57BL/6 mice. We also took advantage of the HEI-OC1 cell line to evaluate the anti-apoptosis effects of PRMT6 knockdown on cisplatin-induced ototoxicity. Apoptotic cells were identified using cleaved caspase-3 staining and TUNEL assay. The levels of reactive oxygen species (ROS) were evaluated by DCFH-DA and cellROX green staining. The mitochondrial membrane potential (ΔΨm) were determined by JC-1, TMRM, and rhodamine 123 staining. Results: We found that EPZ020411 significantly alleviated neomycin- and cisplatin-induced cell apoptosis and increased hair cell survival. Moreover, pretreatment with EPZ020411 could attenuate neomycin- and cisplatin-induced hearing loss in vivo. Mechanistic studies revealed that inhibition of PRMT6 could reverse the increased expression of caspase-3 and cytochrome c translocation, mitochondrial dysfunction, increased accumulation of ROS, and activation of cell apoptosis after cisplatin injury. Conclusions: Our findings suggested that PRMT6 might serve as a new therapeutic target to prevent hearing loss caused by aminoglycoside- and cisplatin-induced ototoxicity by preventing ROS formation and modulating the mitochondria-related damage and apoptosis.


Cisplatin/toxicity , Enzyme Inhibitors/pharmacology , Hair Cells, Auditory/metabolism , Hearing Loss/metabolism , Neomycin/toxicity , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Animals , Anti-Bacterial Agents/toxicity , Apoptosis , Cell Line , Hair Cells, Auditory/pathology , Hearing Loss/chemically induced , Hearing Loss/pathology , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Protein-Arginine N-Methyltransferases/physiology , Reactive Oxygen Species/metabolism
12.
Cell Death Differ ; 27(1): 15-28, 2020 01.
Article En | MEDLINE | ID: mdl-31000813

Cellular senescence is implicated in aging or age-related diseases. Sonic hedgehog (Shh) signaling, an inducer of embryonic development, has recently been demonstrated to inhibit cellular senescence. However, the detailed mechanisms to activate Shh signaling to prevent senescence is not well understood. Here, we demonstrate that Protein arginine methyltransferase 7 (PRMT7) promotes Shh signaling via GLI2 methylation which is critical for suppression of cellular senescence. PRMT7-deficient mouse embryonic fibroblasts (MEFs) exhibited a premature cellular senescence with accompanied increase in the cell cycle inhibitors p16 and p21. PRMT7 depletion results in reduced Shh signaling activity in MEFs while PRMT7 overexpression enhances GLI2-reporter activities that are sensitive to methylation inhibition. PRMT7 interacts with and methylates GLI2 on arginine residues 225 and 227 nearby a binding region of SUFU, a negative regulator of GLI2. This methylation interferes with GLI2-SUFU binding, leading to facilitation of GLI2 nuclear accumulation and Shh signaling. Taken together, these data suggest that PRMT7 induces GLI2 methylation, reducing its binding to SUFU and increasing Shh signaling, ultimately leading to prevention of cellular senescence.


Cellular Senescence , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Zinc Finger Protein Gli2/metabolism , Arginine/metabolism , Cell Nucleus , Cells, Cultured , Cilia/metabolism , Hedgehog Proteins/physiology , Methylation , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/physiology , Repressor Proteins/antagonists & inhibitors , Signal Transduction , Zinc Finger Protein Gli2/chemistry
13.
Cell Biol Int ; 44(3): 882-893, 2020 Mar.
Article En | MEDLINE | ID: mdl-31829474

Accumulating studies have indicated that long non-coding RNAs (lncRNAs) are crucial modulators in cancer biology. In this work, we investigated the function and related mechanisms of LINC01436 in the progression of gastric cancer (GC). We demonstrated that LINC01436 was significantly up-regulated in cancerous tissues of GC samples, and its overexpression was correlated with a worse prognosis for the patients. In the GC cell line BGC823 cells, LINC01436 knockdown repressed the proliferation and metastasis of cancer cells; conversely, in GC cell line AGS cells, overexpression of LINC01436 showed the opposite effects. We then demonstrated that miR-585, a tumor suppressor, could bind to both LINC01436 and the 3'-UTR of F-box protein 11 (FBOX11), and LINC01436 was proved to sponge miR-585 and repress it, and indirectly promoted the expression of FBOX11. Collectively, these results suggested that LINC01436 was an oncogenic lncRNA in GC and promoted proliferation and metastasis of GC cell via regulating miR-585 and FBOX11.


F-Box Proteins/physiology , MicroRNAs/metabolism , Protein-Arginine N-Methyltransferases/physiology , RNA, Long Noncoding/physiology , Stomach Neoplasms/metabolism , Adult , Aged , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Humans , Male , Middle Aged
14.
Nat Struct Mol Biol ; 26(11): 999-1012, 2019 11.
Article En | MEDLINE | ID: mdl-31611688

Protein arginine methyltransferase 5 (PRMT5) has emerged as a promising cancer drug target, and three PRMT5 inhibitors are currently in clinical trials for multiple malignancies. In this study, we investigated the role of PRMT5 in human acute myeloid leukemia (AML). Using an enzymatic dead version of PRMT5 and a PRMT5-specific inhibitor, we demonstrated the requirement of the catalytic activity of PRMT5 for the survival of AML cells. We then identified PRMT5 substrates using multiplexed quantitative proteomics and investigated their role in the survival of AML cells. We found that the function of the splicing regulator SRSF1 relies on its methylation by PRMT5 and that loss of PRMT5 leads to changes in alternative splicing of multiple essential genes. Our study proposes a mechanism for the requirement of PRMT5 for leukemia cell survival and provides potential biomarkers for the treatment response to PRMT5 inhibitors.


Leukemia, Myeloid, Acute/enzymology , Neoplasm Proteins/physiology , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/physiology , RNA Splicing , Animals , Antineoplastic Agents/pharmacology , Arginine/analogs & derivatives , Arginine/biosynthesis , Arginine/metabolism , Catalysis , Cell Line, Tumor , Cell Survival , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/genetics , Methylation , Mice , Molecular Targeted Therapy , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Protein Processing, Post-Translational/genetics , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/deficiency , Proteomics , Serine-Arginine Splicing Factors/metabolism
15.
Arch Toxicol ; 93(8): 2335-2346, 2019 08.
Article En | MEDLINE | ID: mdl-31256211

This study investigated whether protein arginine methyltransferase (PRMT) and the cannabinoid system are involved in cisplatin-induced ototoxicity. Cisplatin increased cytosine-cytosine-adenosine-adenosine-thymidine-enhancer-binding protein homologous protein expression. This effect is indicative of an increase in endoplasmic reticulum (ER) stress, and apoptosis signaling including cleavage of caspase-3, caspase-9, poly-adenosine diphosphate-ribose polymerase, and phospho-p53, as well as expression of PRMT3, PRMT4 and fatty acid amide hydrolase (FAAH)1 in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells. In addition, overexpression of PRMT3 or PRMT4 increased the expression of FAAH1 expression, apoptosis, and ER stress signaling in HEI-OC1 cells, whereas PRMT3 or PRMT4 knockdown had the opposite effect. Furthermore, overexpression of FAAH1 increased apoptosis and ER stress, but expression of the PRMTs was unchanged. In addition, a cannabinoid 1 receptor agonist and FAAH inhibitor attenuated apoptosis and ER stress, while cisplatin increased the binding of PRMT3 with FAAH1. In the in vivo experiments, cisplatin was injected intraperitoneally at 6 mg/kg/day into C57BL/6 mice, and 7 days later, this study confirmed that PRMT3 and PRMT4 were upregulated in the organ of Corti of the mice. These results indicate that cisplatin-induced ototoxicity was correlated with PRMT3, PRMT4 and the cannabinoid system, and PRMT3 binding with FAAH1 was increased by cisplatin in HEI-OC1 cells. Therefore, this study suggests that PRMT3 mediates cisplatin-induced ototoxicity via interaction with FAAH1 in vitro and in vivo.


Cisplatin/toxicity , Ototoxicity/etiology , Protein-Arginine N-Methyltransferases/physiology , Receptor, Cannabinoid, CB1/physiology , Amidohydrolases/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Male , Mice , Mice, Inbred C57BL
17.
Cell Rep ; 26(9): 2316-2328.e6, 2019 02 26.
Article En | MEDLINE | ID: mdl-30811983

Protein arginine methyltransferase 5 (PRMT5) is essential for hematopoiesis, while PRMT5 inhibition remains a promising therapeutic strategy against various cancers. Here, we demonstrate that hematopoietic stem cell (HSC) quiescence and viability are severely perturbed upon PRMT5 depletion, which also increases HSC size, PI3K/AKT/mechanistic target of rapamycin (mTOR) pathway activity, and protein synthesis rate. We uncover a critical role for PRMT5 in maintaining HSC genomic integrity by modulating splicing of genes involved in DNA repair. We found that reducing PRMT5 activity upregulates exon skipping and intron retention events that impair gene expression. Genes across multiple DNA repair pathways are affected, several of which mediate interstrand crosslink repair and homologous recombination. Consequently, loss of PRMT5 activity leads to endogenous DNA damage that triggers p53 activation, induces apoptosis, and culminates in rapid HSC exhaustion, which is significantly delayed by p53 depletion. Collectively, these findings establish the importance of cell-intrinsic PRMT5 activity in HSCs.


Hematopoietic Stem Cells/enzymology , Protein-Arginine N-Methyltransferases/physiology , Proteostasis , RNA Splicing , Animals , Apoptosis , Cell Line , DNA Damage , DNA Repair , Genome , Hematopoietic Stem Cells/metabolism , Mice , Protein Biosynthesis , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
18.
Mol Biol Cell ; 30(6): 778-793, 2019 03 15.
Article En | MEDLINE | ID: mdl-30699057

Protein arginine methyltransferases (PRMTs) are a family of enzymes that modify proteins by methylating the guanidino nitrogen atoms of arginine residues to regulate cellular processes such as chromatin remodeling, pre-mRNA splicing, and signal transduction. PRMT7 is the single type III PRMT solely capable of arginine monomethylation. To date, other than histone proteins, there are very few identified substrates of PRMT7. We therefore performed quantitative mass spectrometry experiments to identify PRMT7's interactome and potential substrates to better characterize the enzyme's biological function(s) in cells. These experiments revealed that PRMT7 interacts with and can methylate eukaryotic translation initiation factor 2 alpha (eIF2α), in vitro and in breast cancer cells. Furthermore, we uncovered a potential regulatory interplay between eIF2α arginine methylation by PRMT7 and stress-induced phosphorylation status of eIF2α at serine 51. Finally, we demonstrated that PRMT7 is required for eIF2α-dependent stress granule formation in the face of various cellular stresses. Altogether, our findings implicate PRMT7 as a novel mediator of eIF2α-dependent cellular stress response pathways.


Eukaryotic Initiation Factor-2/metabolism , Protein-Arginine N-Methyltransferases/physiology , Amino Acid Sequence , Arginine/metabolism , Cell Line , Cytosol/physiology , DNA Methylation , Eukaryotic Initiation Factor-2/physiology , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Histones/metabolism , Humans , MCF-7 Cells , Methylation , Phosphorylation , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , Stress, Physiological/physiology
19.
Nat Commun ; 10(1): 22, 2019 01 03.
Article En | MEDLINE | ID: mdl-30604754

Mechanisms regulating B cell development, activation, education in the germinal center (GC) and differentiation, underpin the humoral immune response. Protein arginine methyltransferase 5 (Prmt5), which catalyzes most symmetric dimethyl arginine protein modifications, is overexpressed in B cell lymphomas but its function in normal B cells is poorly defined. Here we show that Prmt5 is necessary for antibody responses and has essential but distinct functions in all proliferative B cell stages in mice. Prmt5 is necessary for B cell development by preventing p53-dependent and p53-independent blocks in Pro-B and Pre-B cells, respectively. By contrast, Prmt5 protects, via p53-independent pathways, mature B cells from apoptosis during activation, promotes GC expansion, and counters plasma cell differentiation. Phenotypic and RNA-seq data indicate that Prmt5 regulates GC light zone B cell fate by regulating transcriptional programs, achieved in part by ensuring RNA splicing fidelity. Our results establish Prmt5 as an essential regulator of B cell biology.


B-Lymphocytes/physiology , Cell Proliferation/physiology , Germinal Center/physiology , Immunity, Humoral/physiology , Protein-Arginine N-Methyltransferases/physiology , Animals , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Female , Gene Knockdown Techniques , Germinal Center/cytology , Humans , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction/physiology , Trichostrongyloidea/immunology , Trichostrongyloidiasis/immunology , Trichostrongyloidiasis/parasitology , Tumor Suppressor Protein p53/metabolism
20.
Cancer Res ; 79(1): 21-32, 2019 01 01.
Article En | MEDLINE | ID: mdl-30352814

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.


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
...