29 results on '"PRMT6"'
Search Results
2. PRMT6 promotes colorectal cancer progress via activating MYC signaling.
- Author
-
Zhang, Xin, Jin, Mingxin, Chu, Yali, Liu, Fengjun, Qu, Hui, and Chen, Cheng
- Subjects
- *
CANCER cell proliferation , *MEDICAL sciences , *PROTEIN arginine methyltransferases , *POST-translational modification , *COLORECTAL cancer - Abstract
Colorectal cancer (CRC) remains a major global health challenge, with high rates of incidence and mortality. This study investigates the role of protein arginine methyltransferase 6 (PRMT6) as an oncogene in CRC and its mechanistic involvement in tumor progression. We found that PRMT6 is significantly overexpressed in CRC tissues compared to adjacent normal tissues and is associated with poorer patient survival. Functional assays demonstrated that PRMT6 promotes CRC cell proliferation, migration, and invasion. Mechanistically, PRMT6 enhances MYC signaling by stabilizing c-MYC through mono-methylation at arginine 371, which inhibits c-MYC poly-ubiquitination and subsequent degradation. This post-translational modification is crucial for PRMT6-induced cancer cell proliferation. Xenograft models further validated that PRMT6 knockdown results in reduced tumor growth and decreased c-MYC levels. Our findings highlight PRMT6 as a key regulator of c-MYC stability and CRC progression, suggesting that targeting PRMT6 or its effects on c-MYC could offer a promising strategy for CRC treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
3. KLF4 regulates trophoblast function and associates with unexplained recurrent spontaneous abortion
- Author
-
Yiling Tan, Jiayu Wang, Chunming Liu, Shujuan Wu, Mengqi Zhou, Yan Zhang, Tailang Yin, and Jing Yang
- Subjects
KLF4 ,Unexplained recurrent spontaneous abortion ,PRMT6 ,H3R2ME2a ,Trophoblast invasion ,Medicine - Abstract
Abstract Background Recurrent spontaneous abortion (RSA) is defined as two or more consecutive spontaneous abortions before 20 weeks with the same spouse [1]. However, approximately 50% of RSA cases of unknown cause are classified as unexplained recurrent spontaneous abortion (URSA). Potential factors include decreased trophoblast cell migration and invasion, leading to impaired placental implantation and maintenance of the normal maternal-fetal interface. However, the mechanism of this pathogenesis remains unknown. In this study, we investigated the potential role and mechanism of KLF4 in regulating URSA by influencing the invasion and migration ability of trophoblast cells. Methods We firstly identified 817 differentially expressed genes by performing a difference analysis of the dataset GSE121950 [2] related to recurrent abortion, and intersected the top 10 genes obtained respectively by the three algorithms: DMNC, MNC, and EPC using Venn Diagram.To detect the expression levels of core genes, villi samples were obtained from normal pregnant women and patients with URSA. RT-qPCR analysis revealed a significant difference in KLF4 mRNA expression and KLF4 was then analyzed. Trophoblast cell lines HTR8 and JEG3 were used to investigate the effect of KLF4 on trophoblastic function. Wound healing and transwell assays was performed to detect the invasion and migration of trophoblast cells. The expression of epithelial-mesenchymal transition(EMT) molecules were detected by RT-qPCR and western blot. Promoter detection and epigenetic modification were detected by chromatin immunoprecipitation (ChIP) assay. Molecular nuclear localization was detected by immunofluorescence and subcellular fractionation. Miscarried mice model was used to study the effects of KLF4 on URSA induced by reduced trophoblast invasion and migration. Results KLF4 is highly expressed in the villi of patients with URSA. KLF4 inhibits the expression level of H3R2ME2a in trophoblast cells by regulating the transcriptional level and nuclear translocation of PRMT6, thereby inhibiting the possible regulatory mechanism of trophoblastic invasion and providing a potential treatment strategy for URSA in vivo. Conclusions The KLF4/PRMT6/H3R2ME2a axis regulates mechanisms associated with unexplained recurrent spontaneous abortion by regulating trophoblast function.
- Published
- 2024
- Full Text
- View/download PDF
4. PRMT6 Epigenetically Drives Metabolic Switch from Fatty Acid Oxidation toward Glycolysis and Promotes Osteoclast Differentiation During Osteoporosis.
- Author
-
Chu, Wenxiang, Peng, Weilin, Lu, Yingying, Liu, Yishan, Li, Qisheng, Wang, Haibin, Wang, Liang, Zhang, Bangke, Liu, Zhixiao, Han, Lin, Ma, Hongdao, Yang, Haisong, Han, Chaofeng, and Lu, Xuhua
- Subjects
- *
FATTY acid oxidation , *METABOLIC reprogramming , *METABOLIC regulation , *CELL determination , *GLYCOLYSIS - Abstract
Epigenetic regulation of metabolism profoundly influences cell fate commitment. During osteoclast differentiation, the activation of RANK signaling is accompanied by metabolic reprogramming, but the epigenetic mechanisms by which RANK signaling induces this reprogramming remain elusive. By transcriptional sequence and ATAC analysis, this study identifies that activation of RANK signaling upregulates PRMT6 by epigenetic modification, triggering a metabolic switching from fatty acids oxidation toward glycolysis. Conversely, Prmt6 deficiency reverses this shift, markedly reducing HIF‐1α‐mediated glycolysis and enhancing fatty acid oxidation. Consequently, PRMT6 deficiency or inhibitor impedes osteoclast differentiation and alleviates bone loss in ovariectomized (OVX) mice. At the molecular level, Prmt6 deficiency reduces asymmetric dimethylation of H3R2 at the promoters of genes including Ppard, Acox3, and Cpt1a, enhancing genomic accessibility for fatty acid oxidation. PRMT6 thus emerges as a metabolic checkpoint, mediating metabolic switch from fatty acid oxidation to glycolysis, thereby supporting osteoclastogenesis. Unveiling PRMT6's critical role in epigenetically orchestrating metabolic shifts in osteoclastogenesis offers a promising target for anti‐resorptive therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. HSV-1 reactivation results in post-herpetic neuralgia by upregulating Prmt6 and inhibiting cGAS-STING.
- Author
-
Kong, Erliang, Hua, Tong, Li, Jian, Li, Yongchang, Yang, Mei, Ding, Ruifeng, Wang, Haowei, Wei, Huawei, Feng, Xudong, Han, Chaofeng, and Yuan, Hongbin
- Subjects
- *
HUMAN herpesvirus 1 , *METHYLTRANSFERASES , *PROTEIN arginine methyltransferases , *INTERFERON regulatory factors , *TYPE I interferons , *NEURALGIA - Abstract
Chronic varicella zoster virus (VZV) infection induced neuroinflammatory condition is the critical pathology of post-herpetic neuralgia (PHN). The immune escape mechanism of VZV remains elusive. As to mice have no VZV infection receptor, herpes simplex virus type 1 (HSV-1) infection is a well established PHN mice model. Transcriptional expression analysis identified that the protein arginine methyltransferases 6 (Prmt6) was upregulated upon HSV-1 infection, which was further confirmed by immunofluorescence staining in spinal dorsal horn. Prmt6 deficiency decreased HSV-1-induced neuroinflammation and PHN by enhancing antiviral innate immunity and decreasing HSV-1 load in vivo and in vitro. Overexpression of Prmt6 in microglia dampened antiviral innate immunity and increased HSV-1 load. Mechanistically, Prmt6 methylated and inactivated STING, resulting in reduced phosphorylation of TANK binding kinase-1 (TBK1) and interferon regulatory factor 3 (IRF3), diminished production of type I interferon (IFN-I) and antiviral innate immunity. Furthermore, intrathecal or intraperitoneal administration of the Prmt6 inhibitor EPZ020411 decreased HSV-1-induced neuroinflammation and PHN by enhancing antiviral innate immunity and decreasing HSV-1 load. Our findings revealed that HSV-1 escapes antiviral innate immunity and results in PHN by upregulating Prmt6 expression and inhibiting the cGAS-STING pathway, providing novel insights and a potential therapeutic target for PHN. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. PRMT6-mediated transcriptional activation of ythdf2 promotes glioblastoma migration, invasion, and emt via the wnt–β-catenin pathway
- Author
-
Peng Yu, Tutu Xu, Wenmeng Ma, Xiang Fang, Yue Bao, Chengran Xu, Jinhai Huang, Yongqing Sun, and Guangyu Li
- Subjects
GBM ,PRMT6 ,Migration ,Invasion ,EMT ,YTHDF2 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Protein arginine methyltransferase 6 (PRMT6) plays a crucial role in various pathophysiological processes and diseases. Glioblastoma (GBM; WHO Grade 4 glioma) is the most common and lethal primary brain tumor in adults, with a prognosis that is extremely poor, despite being less common than other systemic malignancies. Our current research finds PRMT6 upregulated in GBM, enhancing tumor malignancy. Yet, the specifics of PRMT6’s regulatory processes and potential molecular mechanisms in GBM remain largely unexplored. Methods PRMT6’s expression and prognostic significance in GBM were assessed using glioma public databases, immunohistochemistry (IHC), and immunoblotting. Scratch and Transwell assays examined GBM cell migration and invasion. Immunoblotting evaluated the expression of epithelial-mesenchymal transition (EMT) and Wnt-β-catenin pathway-related proteins. Dual-luciferase reporter assays and ChIP-qPCR assessed the regulatory relationship between PRMT6 and YTHDF2. An in situ tumor model in nude mice evaluated in vivo conditions. Results Bioinformatics analysis indicates high expression of PRMT6 and YTHDF2 in GBM, correlating with poor prognosis. Functional experiments show PRMT6 and YTHDF2 promote GBM migration, invasion, and EMT. Mechanistic experiments reveal PRMT6 and CDK9 co-regulate YTHDF2 expression. YTHDF2 binds and promotes the degradation of negative regulators APC and GSK3β mRNA of the Wnt-β-catenin pathway, activating it and consequently enhancing GBM malignancy. Conclusions Our results demonstrate the PRMT6-YTHDF2-Wnt-β-Catenin axis promotes GBM migration, invasion, and EMT in vitro and in vivo, potentially serving as a therapeutic target for GBM.
- Published
- 2024
- Full Text
- View/download PDF
7. PRMT6-mediated transcriptional activation of ythdf2 promotes glioblastoma migration, invasion, and emt via the wnt–β-catenin pathway.
- Author
-
Yu, Peng, Xu, Tutu, Ma, Wenmeng, Fang, Xiang, Bao, Yue, Xu, Chengran, Huang, Jinhai, Sun, Yongqing, and Li, Guangyu
- Subjects
BRAIN tumors ,PROTEIN arginine methyltransferases ,GLIOBLASTOMA multiforme ,EPITHELIAL-mesenchymal transition ,GLIOMAS ,CELL migration - Abstract
Background: Protein arginine methyltransferase 6 (PRMT6) plays a crucial role in various pathophysiological processes and diseases. Glioblastoma (GBM; WHO Grade 4 glioma) is the most common and lethal primary brain tumor in adults, with a prognosis that is extremely poor, despite being less common than other systemic malignancies. Our current research finds PRMT6 upregulated in GBM, enhancing tumor malignancy. Yet, the specifics of PRMT6's regulatory processes and potential molecular mechanisms in GBM remain largely unexplored. Methods: PRMT6's expression and prognostic significance in GBM were assessed using glioma public databases, immunohistochemistry (IHC), and immunoblotting. Scratch and Transwell assays examined GBM cell migration and invasion. Immunoblotting evaluated the expression of epithelial-mesenchymal transition (EMT) and Wnt-β-catenin pathway-related proteins. Dual-luciferase reporter assays and ChIP-qPCR assessed the regulatory relationship between PRMT6 and YTHDF2. An in situ tumor model in nude mice evaluated in vivo conditions. Results: Bioinformatics analysis indicates high expression of PRMT6 and YTHDF2 in GBM, correlating with poor prognosis. Functional experiments show PRMT6 and YTHDF2 promote GBM migration, invasion, and EMT. Mechanistic experiments reveal PRMT6 and CDK9 co-regulate YTHDF2 expression. YTHDF2 binds and promotes the degradation of negative regulators APC and GSK3β mRNA of the Wnt-β-catenin pathway, activating it and consequently enhancing GBM malignancy. Conclusions: Our results demonstrate the PRMT6-YTHDF2-Wnt-β-Catenin axis promotes GBM migration, invasion, and EMT in vitro and in vivo, potentially serving as a therapeutic target for GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Arginine methylation of ALKBH5 by PRMT6 promotes breast tumorigenesis via LDHA-mediated glycolysis.
- Author
-
Han, Xue, Ren, Chune, Jiang, Aifang, Sun, Yonghong, Lu, Jiayi, Ling, Xi, Lu, Chao, and Yu, Zhenhai
- Abstract
ALKBH5 is a master regulator of N6-methyladenosine (m
6 A) modification, which plays a crucial role in many biological processes. Here, we show that ALKBH5 is required for breast tumor growth. Interestingly, PRMT6 directly methylates ALKBH5 at R283, which subsequently promotes breast tumor growth. Furthermore, arginine methylation of ALKBH5 by PRMT6 increases LDHA RNA stability via m6 A demethylation, leading to increased aerobic glycolysis. Moreover, PRMT6-mediated ALKBH5 arginine methylation is confirmed in PRMT6-knockout mice. Collectively, these findings identify a PRMT6-ALKBH5-LDHA signaling axis as a novel target for the treatment of breast cancer. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
9. Machine Learning for Digital Scoring of PRMT6 in Immunohistochemical Labeled Lung Cancer.
- Author
-
Mahmoud, Abeer M., Brister, Eileen, David, Odile, Valyi-Nagy, Klara, Sverdlov, Maria, Gann, Peter H., and Kim, Sage J.
- Subjects
- *
IMMUNOHISTOCHEMISTRY , *MACHINE learning , *LUNG tumors , *EARLY detection of cancer , *GENE expression , *INTRACLASS correlation , *RESEARCH funding , *TUMOR markers , *STATISTICAL correlation , *LONGITUDINAL method - Abstract
Simple Summary: We trained and validated the machine learning digital scoring method of PRMT6 protein expression in lung cancer tissue samples. PRMT6 is an important biomarker for the progression of lung cancer; however, conventional pathologists' manual scoring of its expression in large samples is time consuming, particularly when analyzing large sections of lung cancer tissue. Using HALO software, we optimized the digital method for scoring PRMT6 expression on immunohistochemically stained lung cancer tissue. Our optimized digital scoring showed excellent concordance with two pathologists using the immunoreactive scoring method. Our findings showed that digital scoring trained by pathologists can be a more efficient method with a high level of accuracy. Lung cancer is the leading cause of cancer death in the U.S. Therefore, it is imperative to identify novel biomarkers for the early detection and progression of lung cancer. PRMT6 is associated with poor lung cancer prognosis. However, analyzing PRMT6 expression manually in large samples is time-consuming posing a significant limitation for processing this biomarker. To overcome this issue, we trained and validated an automated method for scoring PRMT6 in lung cancer tissues, which can then be used as the standard method in future larger cohorts to explore population-level associations between PRMT6 expression and sociodemographic/clinicopathologic characteristics. We evaluated the ability of a trained artificial intelligence (AI) algorithm to reproduce the PRMT6 immunoreactive scores obtained by pathologists. Our findings showed that tissue segmentation to cancer vs. non-cancer tissues was the most critical parameter, which required training and adjustment of the algorithm to prevent scoring non-cancer tissues or ignoring relevant cancer cells. The trained algorithm showed a high concordance with pathologists with a correlation coefficient of 0.88. The inter-rater agreement was significant, with an intraclass correlation of 0.95 and a scale reliability coefficient of 0.96. In conclusion, we successfully optimized a machine learning algorithm for scoring PRMT6 expression in lung cancer that matches the degree of accuracy of scoring by pathologists. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
10. Arginine Methylation of Integrin Alpha-4 Prevents Fibrosis Development in Alcohol-Associated Liver DiseaseSummary
- Author
-
Michael Schonfeld, Maria T. Villar, Antonio Artigues, Steven A. Weinman, and Irina Tikhanovich
- Subjects
PRMT6 ,ALD ,Inflammation ,TGFβ ,Macrophage ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: Alcohol-associated liver disease (ALD) comprises a spectrum of disorders including steatosis, steatohepatitis, fibrosis, and cirrhosis. We aimed to study the role of protein arginine methyltransferase 6 (PRMT6), a new regulator of liver function, in ALD progression. Methods: Prmt6-deficient mice and wild-type littermates were fed Western diet with alcohol in the drinking water for 16 weeks. Mice fed standard chow diet or Western diet alone were used as a control. Results: We found that PRMT6 expression in the liver is down-regulated in 2 models of ALD and negatively correlates with disease severity in mice and human liver specimens. Prmt6-deficient mice spontaneously developed liver fibrosis after 1 year and more advanced fibrosis after high-fat diet feeding or thioacetamide treatment. In the presence of alcohol Prmt6 deficiency resulted in a dramatic increase in fibrosis development but did not affect lipid accumulation or liver injury. In the liver PRMT6 is primarily expressed in macrophages and endothelial cells. Transient replacement of knockout macrophages with wild-type macrophages in Prmt6 knockout mice reduced profibrotic signaling and prevented fibrosis progression. We found that PRMT6 decreases profibrotic signaling in liver macrophages via methylation of integrin α-4 at R464 residue. Integrin α-4 is predominantly expressed in infiltrating monocyte derived macrophages. Blocking monocyte infiltration into the liver with CCR2 inhibitor reduced fibrosis development in knockout mice and abolished differences between genotypes. Conclusions: Taken together, our data suggest that alcohol-mediated loss of Prmt6 contributes to alcohol-associated fibrosis development through reduced integrin methylation and increased profibrotic signaling in macrophages.
- Published
- 2023
- Full Text
- View/download PDF
11. PRMT6 promotes tumorigenicity and cisplatin response of lung cancer through triggering 6PGD/ENO1 mediated cell metabolism
- Author
-
Mingming Sun, Leilei Li, Yujia Niu, Yingzhi Wang, Qi Yan, Fei Xie, Yaya Qiao, Jiaqi Song, Huanran Sun, Zhen Li, Sizhen Lai, Hongkai Chang, Han Zhang, Jiyan Wang, Chenxin Yang, Huifang Zhao, Junzhen Tan, Yanping Li, Shuangping Liu, Bin Lu, Min Liu, Guangyao Kong, Yujun Zhao, Chunze Zhang, Shu-Hai Lin, Cheng Luo, Shuai Zhang, and Changliang Shan
- Subjects
Lung cancer ,Metabolic reprogramming ,Post-translational modification ,PRMT6 ,Pentose phosphate pathway flux ,Glycolysis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Metabolic reprogramming is a hallmark of cancer, including lung cancer. However, the exact underlying mechanism and therapeutic potential are largely unknown. Here we report that protein arginine methyltransferase 6 (PRMT6) is highly expressed in lung cancer and is required for cell metabolism, tumorigenicity, and cisplatin response of lung cancer. PRMT6 regulated the oxidative pentose phosphate pathway (PPP) flux and glycolysis pathway in human lung cancer by increasing the activity of 6-phospho-gluconate dehydrogenase (6PGD) and α-enolase (ENO1). Furthermore, PRMT6 methylated R324 of 6PGD to enhancing its activity; while methylation at R9 and R372 of ENO1 promotes formation of active ENO1 dimers and 2-phosphoglycerate (2-PG) binding to ENO1, respectively. Lastly, targeting PRMT6 blocked the oxidative PPP flux, glycolysis pathway, and tumor growth, as well as enhanced the anti-tumor effects of cisplatin in lung cancer. Together, this study demonstrates that PRMT6 acts as a post-translational modification (PTM) regulator of glucose metabolism, which leads to the pathogenesis of lung cancer. It was proven that the PRMT6-6PGD/ENO1 regulatory axis is an important determinant of carcinogenesis and may become a promising cancer therapeutic strategy.
- Published
- 2023
- Full Text
- View/download PDF
12. Asymmetric dimethylation of AMPKα1 by PRMT6 contributes to the formation of phase-separated puncta.
- Author
-
Yoo, Yeon Ju, Cho, Giseong, Kim, Dana, Kim, Yoonkyung, Yun, Nuri, and Oh, Young J.
- Subjects
- *
PROTEIN arginine methyltransferases , *UBIQUITINATION , *AMP-activated protein kinases , *PROTEIN kinases , *ENERGY metabolism , *PHASE separation - Abstract
AMP-activated protein kinase (AMPK) is a heterotrimeric serine/threonine kinase comprising α, β, and γ subunits. AMPK is involved in intracellular energy metabolism and functions as a switch that turns various biological pathways in eukaryotes on and off. Several post-translational modifications regulating AMPK function have been demonstrated, including phosphorylation, acetylation, and ubiquitination; however, arginine methylation has not been reported in AMPKα1. We investigated whether arginine methylation occurs in AMPKα1. Screening experiments revealed arginine methylation of AMPKα1 mediated by protein arginine methyltransferase 6 (PRMT6). In vitro methylation and co-immunoprecipitation assays indicated that PRMT6 can directly interact with and methylate AMPKα1 without involvement of other intracellular components. In vitro methylation assays with truncated and point mutants of AMPKα1 revealed that Arg403 is the residue methylated by PRMT6. Immunocytochemical studies showed that the number of AMPKα1 puncta was enhanced in saponin-permeabilized cells when AMPKα1 was co-expressed with PRMT6, suggesting that PRMT6-mediated methylation of AMPKα1 at Arg403 alters the physiological characteristics of AMPKα1 and may lead to liquid-liquid phase separation. [Display omitted] • Protein arginine methyltransferase 6 (PRMT6) directly methylates AMPKα1. • Arginine 403 of AMPKα1 is the residue methylated by PRMT6. • This event may contribute to the formation of the phase-separated puncta. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. The PRMT6/PARP1/CRL4B Complex Regulates the Circadian Clock and Promotes Breast Tumorigenesis.
- Author
-
Yang, Tianshu, Huang, Wei, Ma, Tianyu, Yin, Xin, Zhang, Jingyao, Huo, Miaomiao, Hu, Ting, Gao, Tianyang, Liu, Wei, Zhang, Die, Yu, Hefen, Teng, Xu, Zhang, Min, Qin, Hao, Yang, Yunkai, Yuan, Baowen, and Wang, Yan
- Subjects
- *
CIRCADIAN rhythms , *CLOCK genes , *BREAST , *METASTATIC breast cancer , *MOLECULAR clock , *PROTEIN arginine methyltransferases , *GENE expression , *UBIQUITIN ligases - Abstract
Circadian rhythms, as physiological systems with self‐regulatory functions in living organisms, are controlled by core clock genes and are involved in tumor development. The protein arginine methyltransferase 6 (PRMT6) serves as an oncogene in a myriad of solid tumors, including breast cancer. Hence, the primary aim of the current study is to investigate the molecular mechanisms by which the PRMT6 complex promotes breast cancer progression. The results show that PRMT6, poly(ADP‐ribose) polymerase 1 (PARP1), and the cullin 4 B (CUL4B)‐Ring E3 ligase (CRL4B) complex interact to form a transcription‐repressive complex that co‐occupies the core clock gene PER3 promoter. Moreover, genome‐wide analysis of PRMT6/PARP1/CUL4B targets identifies a cohort of genes that is principally involved in circadian rhythms. This transcriptional‐repression complex promotes the proliferation and metastasis of breast cancer by interfering with circadian rhythm oscillation. Meanwhile, the PARP1 inhibitor Olaparib enhances clock gene expression, thus, reducing breast carcinogenesis, indicating that PARP1 inhibitors have potential antitumor effects in high‐PRMT6 expression breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. PRMT6 promotes tumorigenicity and cisplatin response of lung cancer through triggering 6PGD/ENO1 mediated cell metabolism.
- Author
-
Sun, Mingming, Li, Leilei, Niu, Yujia, Wang, Yingzhi, Yan, Qi, Xie, Fei, Qiao, Yaya, Song, Jiaqi, Sun, Huanran, Li, Zhen, Lai, Sizhen, Chang, Hongkai, Zhang, Han, Wang, Jiyan, Yang, Chenxin, Zhao, Huifang, Tan, Junzhen, Li, Yanping, Liu, Shuangping, and Lu, Bin
- Subjects
CELL metabolism ,LUNG cancer ,PENTOSE phosphate pathway ,CISPLATIN ,PROTEIN arginine methyltransferases ,GLUCOSE-6-phosphate dehydrogenase deficiency - Abstract
Metabolic reprogramming is a hallmark of cancer, including lung cancer. However, the exact underlying mechanism and therapeutic potential are largely unknown. Here we report that protein arginine methyltransferase 6 (PRMT6) is highly expressed in lung cancer and is required for cell metabolism, tumorigenicity, and cisplatin response of lung cancer. PRMT6 regulated the oxidative pentose phosphate pathway (PPP) flux and glycolysis pathway in human lung cancer by increasing the activity of 6-phospho-gluconate dehydrogenase (6PGD) and α -enolase (ENO1). Furthermore, PRMT6 methylated R324 of 6PGD to enhancing its activity; while methylation at R9 and R372 of ENO1 promotes formation of active ENO1 dimers and 2-phosphoglycerate (2-PG) binding to ENO1, respectively. Lastly, targeting PRMT6 blocked the oxidative PPP flux, glycolysis pathway, and tumor growth, as well as enhanced the anti-tumor effects of cisplatin in lung cancer. Together, this study demonstrates that PRMT6 acts as a post-translational modification (PTM) regulator of glucose metabolism, which leads to the pathogenesis of lung cancer. It was proven that the PRMT6-6PGD/ENO1 regulatory axis is an important determinant of carcinogenesis and may become a promising cancer therapeutic strategy. PRMT6 promotes lung cancer growth and cisplatin response through enhancing oxidative pentose phosphate pathway flux and glycolysis pathway, which mediated by regulating the asymmetric arginine demethylation modification of 6PGD/ENO1. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. PRMT6/LMNA/CXCL12 signaling pathway regulated the osteo/odontogenic differentiation ability in dental stem cells isolated from apical papilla.
- Author
-
Wang, Ning, Li, Miao, Cao, Yangyang, Yang, Haoqing, Li, Le, Ge, Lihua, Fan, Zhipeng, Zhang, Chen, and Jin, Luyuan
- Subjects
- *
STEM cells , *CYTOSKELETAL proteins , *CELLULAR signal transduction , *NUCLEAR proteins , *STROMAL cell-derived factor 1 , *AMELOBLASTS , *BONE regeneration - Abstract
Tooth loss and maxillofacial bone defect are common diseases, which seriously affect people's health. Effective tooth and maxillofacial bone tissue regeneration is a key problem that need to be solved. In the present study, we investigate the role of PRMT6 in osteo/odontogenic differentiation and migration capacity by using SCAPs. Our results showed that knockdown of PRMT6 promoted the osteo/odontogenic differentiation compared with the control group, as detected by alkaline phosphatase activity, alizarin red staining, and the indicators of osteo/odontogenic differentiation measured by Western blot. In addition, overexpression of PRMT6 inhibited the osteo/odontogenic differentiation potentials of SCAPs. Then, knockdown of PRMT6 promoted the migration ability and overexpression of PRMT6 inhibited the migration ability in SCAPs. Mechanically, we discovered that the depletion of PRMT6 promoted the expression of CXCL12 by decreasing H3R2 methylation in the promoter region of CXCL12. In addition, PRMT6 formed a protein complex with LMNA, a nuclear structural protein. Depletion of LMNA inhibited the osteo/odontogenic differentiation and CXCL12 expression and increased the intranucleus PRMT6 in SCAPs. To sum up, PRMT6 might inhibit the osteo/odontogenic differentiation and migration ability of SCAPs via inhibiting CXCL12. And LMNA might be a negative regulator of PRMT6. It is suggested that PRMT6 may be a key target for SCAP-mediated bone and tooth tissue regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
16. This title is unavailable for guests, please login to see more information.
- Author
-
HONDA SHINNOSUKE and HONDA SHINNOSUKE
- Published
- 2024
17. The Emerging Role of PRMT6 in Cancer.
- Author
-
Chen, Zhixian, Gan, Jianfeng, Wei, Zhi, Zhang, Mo, Du, Yan, Xu, Congjian, and Zhao, Hongbo
- Subjects
ALTERNATIVE RNA splicing ,GENETIC regulation ,PROTEIN arginine methyltransferases ,CELLULAR aging ,DNA repair ,METHYLGUANINE ,METHYLTRANSFERASES - Abstract
Protein arginine methyltransferase 6 (PRMT6) is a type I PRMT that is involved in epigenetic regulation of gene expression through methylating histone or non-histone proteins, and other processes such as alternative splicing, DNA repair, cell proliferation and senescence, and cell signaling. In addition, PRMT6 also plays different roles in various cancers via influencing cell growth, migration, invasion, apoptosis, and drug resistant, which make PRMT6 an anti-tumor therapeutic target for a variety of cancers. As a result, many PRMT6 inhibitors are being utilized to explore their efficacy as potential drugs for various cancers. In this review, we summarize the current knowledge on the function and structure of PRMT6. At the same time, we highlight the role of PRMT6 in different cancers, including the differentiation of its promotive or inhibitory effects and the underlying mechanisms. Apart from the above, current research progress and the potential mechanisms of PRMT6 behind them were also summarized. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. Inhibition of PRMT6 reduces neomycin-induced inner ear hair cell injury through the restraint of FoxG1 arginine methylation.
- Author
-
Li, Xingcheng and Chen, Xi
- Subjects
- *
HAIR cells , *INNER ear , *ARGININE , *PROTEIN arginine methyltransferases , *METHYLATION , *REACTIVE oxygen species - Abstract
Objective: Hair cells in the inner ear have been demonstrated to be sensitive to the ototoxicity from some beneficial pharmaceutical drugs. This study aimed to explore the role of protein arginine methyltransferase 6 (PRMT6) in the process of neomycin-induced hearing loss and the underlying mechanism. Methods: The neomycin-induced hearing loss mouse model and hair cell injury in vitro model were established. We took advantage of the HEI-OC1 cell line to evaluate PRMT6 expression in neomycin-induced hair cells, and the effect of PRMT6 on mitochondrial function and FoxG1 arginine methylation. Apoptotic cells were assessed and apoptotic marker cleaved caspase-3 level was detected. Reactive oxygen species (ROS) level and mitochondrial membrane potential (MMP) were subsequently measured. Result: The result showed that PRMT6 was significantly upregulated in neomycin-induced HEI-OC-1 cells, and PRMT6 silencing prevented MMP loss, reduced ROS production, as well as decreased cell apoptosis under neomycin treatment. Further results showed that FoxG1 was downregulated in neomycin-induced HEI-OC-1 cells, and PRMT6 promoted the FoxG1-mediated luciferase activity, while PRMT6 silencing reversed this effect. Mechanistic experiments revealed that PRMT6 silencing reduced the arginine methylation level of FoxG1 protein. In vivo, neomycin-induced upregulation of hearing thresholds and increased cell apoptosis, whereas PRMT6 inhibitor partly reversed these effects. Conclusion: Our findings suggested that inhibition of PRMT6 reduced neomycin-induced inner ear hair cell injury through the restraint of FoxG1 arginine methylation. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
19. The Emerging Role of PRMT6 in Cancer
- Author
-
Zhixian Chen, Jianfeng Gan, Zhi Wei, Mo Zhang, Yan Du, Congjian Xu, and Hongbo Zhao
- Subjects
PRMT6 ,arginine methylation ,cancers ,regulation ,PRMT6 inhibitors ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Protein arginine methyltransferase 6 (PRMT6) is a type I PRMT that is involved in epigenetic regulation of gene expression through methylating histone or non-histone proteins, and other processes such as alternative splicing, DNA repair, cell proliferation and senescence, and cell signaling. In addition, PRMT6 also plays different roles in various cancers via influencing cell growth, migration, invasion, apoptosis, and drug resistant, which make PRMT6 an anti-tumor therapeutic target for a variety of cancers. As a result, many PRMT6 inhibitors are being utilized to explore their efficacy as potential drugs for various cancers. In this review, we summarize the current knowledge on the function and structure of PRMT6. At the same time, we highlight the role of PRMT6 in different cancers, including the differentiation of its promotive or inhibitory effects and the underlying mechanisms. Apart from the above, current research progress and the potential mechanisms of PRMT6 behind them were also summarized.
- Published
- 2022
- Full Text
- View/download PDF
20. Regulation of hepatic lipogenesis by asymmetric arginine methylation.
- Author
-
Han, Hye-Sook, Choi, Byeong Hun, Jang, Seo Young, Choi, Seri, Hwang, Geum-Sook, and Koo, Seung-Hoi
- Subjects
LIPID synthesis ,NON-alcoholic fatty liver disease ,TRANSCRIPTION factors ,ARGININE ,LIPID metabolism ,LUCIFERASES ,HOMEOSTASIS - Abstract
Hepatic lipogenesis is elevated in nutrient abundant conditions to convert the excess carbohydrate into triacylglycerol (TAG). Fatty acyl moiety of TAG is eventually transported into adipose tissues by very low density lipoprotein, leading to the accumulation of TAG as a preferred storage form of excess energy. Disruption of the balance between TAG clearance and synthesis leads to the accumulation of lipids in the liver, leading to the progression of non-alcoholic fatty liver disease (NAFLD) including non-alcoholic steatohepatitis. Protein arginine methyltransferase (PRMT) 6 has been linked to the various metabolic processes including hepatic gluconeogenesis, muscle atrophy and lipodystrophy in mouse models. However, the role of PRMT6 in the control of hepatic lipogenesis has not been elucidated to date. We assessed the interaction between PRMT6 and LXR alpha by using co-immunoprecipitation assay. The specific arginine residue of LXR alpha that is methylated by PRMT6 was assessed by LC-MS/MS assay and the functional consequences of LXR alpha methylation was explored by mSREBP-1c luciferase assay. The effect of PRMT6 on hepatic lipogenesis was assessed by adenovirus-mediated ectopic expression of PRMT6 or knockdown of PRMT6 via shRNA in hepatocytes. Finally, the role of PRMT6 in hepatic lipid metabolism in vivo was explored by either ectopic expression of LXR alpha mutant that is defective in PRMT6-mediated arginine methylation or knockdown of PRMT6 in liver. We found that promoter activity of sterol regulatory element binding protein (SREBP) 1c is robustly activated by PRMT6. Interestingly, we demonstrated that PRMT6 binds to LXR alpha, a transcription factor for SREBP-1c, via its LXXLL motif, leading to the asymmetric dimethylation of an arginine residue and activation of this protein. Indeed, ectopic expression of PRMT6 in hepatocytes led to the enhanced expression of LXR alpha target genes in the lipogenic pathway. Conversely, genetic or pharmacological inhibition of PRMT6 diminished expression of lipogenic genes and the lipid accumulation in primary hepatocytes. Mechanistically, we found that asymmetric dimethylation of LXR alpha led to the dissociation of small heterodimer partner (SHP), a transcriptional co-inhibitor of this factor, resulting in the activation of LXR alpha-mediated transcriptional process. Finally, we showed that disruption of asymmetric dimethylation of LXR alpha in the liver led to the diminished expression of genes in the lipogenesis, resulting in the reduced hepatic lipid accumulation in high fat diet-fed mice in vivo. We showed that PRMT6 modulates LXR alpha activity by conferring asymmetric dimethylation of arginine 253, thus blocking SHP-mediated inhibition and promoting hepatic lipid accumulation. These results suggest that PRMT6 is critical in the control of lipid homeostasis by regulation of LXR alpha-mediated lipogenesis in the liver. [Display omitted] • PRMT6 promotes LXR alpha-dependent hepatic lipogenesis. • Arginine 253 methylation is critical in PRMT6-dependent activation of LXR alpha. • PRMT6 blocks recruitment of transcriptional co-repressor to LXR alpha. • Arginine methylation on LXR alpha is critical in hepatic lipid metabolism in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Discovery of a first-in-class degrader for the protein arginine methyltransferase 6 (PRMT6).
- Author
-
Yang, Hongling, Zhang, Qiangsheng, Zhou, Shuyan, Hu, Zuli, Tang, Qing, Li, Zulong, Feng, Qiang, and Yu, Luoting
- Subjects
- *
PROTEIN arginine methyltransferases , *DRUG development , *SMALL molecules , *PROTEINS , *CELL cycle , *METHYLTRANSFERASES - Abstract
[Display omitted] • A series of novel adamantane hydrophobic tags derivatives have been designed and synthesized. • SKLB0124 was developed as the first-in-class degrader for PRMT6. • SKLB-0124 selectively degraded PRMT6 over PRMT1/8. • SKLB-0124 showed significant antiproliferative activities. PRMT6 is a member of the protein arginine methyltransferase family, which participates in a variety of physical processes and plays an important role in the occurrence and development of tumors. Using small molecules to design and synthesize targeted protein degraders is a new strategy for drug development. Here, we report the first-in-class degrader SKLB-0124 for PRMT6 based on the hydrophobic tagging (HyT) method. Importantly, SKLB-0124 induced proteasome dependent degradation of PRMT6 and significantly inhibited the proliferation of HCC827 and MDA-MB-435 cells. Moreover, SKLB-0124 effectively induced apoptosis and cell cycle arrest in these two cell lines. Our data clarified that SKLB-0124 is a promising selective PRMT6 degrader for cancer therapy which is worthy of further evaluation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. PRMT6-mediated ADMA promotes p62 phase separation to form a negative feedback loop in ferroptosis.
- Author
-
Feng L, Chen L, Wang W, Wei Q, Chen M, Jiang X, Hu S, Wu Y, Duan L, Zhu L, Wang X, and Jin H
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Feedback, Physiological, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Pancreatic Neoplasms genetics, Sequestosome-1 Protein metabolism, Sequestosome-1 Protein genetics, Mice, Nude, Signal Transduction, Phase Separation, RNA-Binding Proteins, Ferroptosis, Protein-Arginine N-Methyltransferases metabolism, Protein-Arginine N-Methyltransferases genetics, Arginine metabolism, Arginine analogs & derivatives, NF-E2-Related Factor 2 metabolism, NF-E2-Related Factor 2 genetics, Kelch-Like ECH-Associated Protein 1 metabolism, Kelch-Like ECH-Associated Protein 1 genetics
- Abstract
Purpose: Due to intrinsic defensive response, ferroptosis-activating targeted therapy fails to achieve satisfactory clinical benefits. Though p62-Keap1-Nrf2 axis is activated to form a negative feedback loop during ferroptosis induction, how p62 is activated remains largely unknown. Methods: MTS assay was applied to measure cell growth. Lipid ROS was detected with C11-BODIPY reagent by flow cytometer. Quantitative real-time PCR (qPCR) and western blotting were performed to determine mRNA and protein level. Immunofluorescence (IF) was performed to examine the distribution of proteins. Fluorescence recovery after photobleaching (FRAP) was adopted to evaluate p62 phase separation. Immunoprecipitation (IP), co-IP and Proximal ligation assay (PLA) were performed to detected protein posttranslational modifications and protein-protein interactions. Tumor xenograft model was employed to inspect in vivo growth of pancreatic cancer cells. Results: Upon ferroptosis induction, Nuclear Factor E2 Related Factor 2 (Nrf2) protein and its downstream genes such as HMOX1 and NQO1 were upregulated. Knockdown of p62 significantly reversed Nrf2 upregulation and Keap1 decrease after ferroptosis induction. Knockdown of either p62 or Nrf2 remarkably sensitized ferroptosis induction. Due to augmented p62 phase separation, formation of p62 bodies were increased to recruit Keap1 after ferroptosis induction. Protein arginine methyltransferase 6 (PRMT6) mediated asymmetric dimethylarginine (ADMA) of p62 to increase its oligomerization, promoting p62 phase separation and p62 body formation. Knockdown of p62 or PRMT6 notably sensitized pancreatic cancer cells to ferroptosis both in vitro and in vivo through suppressing Nrf2 signaling. Conclusion: During ferroptosis induction, PRMT6 mediated p62 ADMA to promote its phase separation, sequestering Keap1 to activate Nrf2 signaling and inhibit ferroptosis. Therefore, targeting PRMT6-mediated p62 ADMA could be a new option to sensitize ferroptosis for cancer treatment., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2024
- Full Text
- View/download PDF
23. Chimeric PRMT6 protein produced by an endogenous retrovirus promoter regulates cell fate decision in mouse preimplantation embryos†.
- Author
-
Honda S, Hatamura M, Kunimoto Y, Ikeda S, and Minami N
- Subjects
- Animals, Mice, Leucine metabolism, Methylation, Histones genetics, Histones metabolism, Blastocyst metabolism, Arginine, Recombinant Fusion Proteins genetics, RNA, Transfer metabolism, Mammals genetics, Endogenous Retroviruses genetics
- Abstract
Murine endogenous retrovirus with leucine tRNA primer, also known as MERVL, is expressed during zygotic genome activation in mammalian embryos. Here we show that protein arginine N-methyltransferase 6 (Prmt6) forms a chimeric transcript with MT2B2, one of the long terminal repeat sequences of murine endogenous retrovirus with leucine tRNA primer, and is translated into an elongated chimeric protein (PRMT6MT2B2) whose function differs from that of the canonical PRMT6 protein (PRMT6CAN) in mouse preimplantation embryos. Overexpression of PRMT6CAN in fibroblast cells increased asymmetric dimethylation of the third arginine residue of both histone H2A (H2AR3me2a) and histone H4 (H4R3me2a), while overexpression of PRMT6MT2B2 increased only H2AR3me2a. In addition, overexpression of PRMT6MT2B2 in one blastomere of mouse two-cell embryos promoted cell proliferation and differentiation of the blastomere into epiblast cells at the blastocyst stage, while overexpression of PRMT6CAN repressed cell proliferation. This is the first report of the translation of a chimeric protein (PRMT6MT2B2) in mouse preimplantation embryos. Our results suggest that analyzing chimeric transcripts with murine endogenous retrovirus with leucine tRNA primer will provide insight into the relationship between zygotic genome activation and subsequent intra- and extra-cellular lineage determination., (© The Author(s) 2024. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
24. Inhibition of protein arginine methyltransferase 6 activates interferon signaling and induces the apoptosis of endometrial cancer cells via histone modification.
- Author
-
Inoue F, Sone K, Kumegawa K, Hachijo R, Suzuki E, Tanimoto S, Tsuboyama N, Kato K, Toyohara Y, Takahashi Y, Kusakabe M, Kukita A, Honjoh H, Nishijima A, Taguchi A, Miyamoto Y, Tanikawa M, Iriyama T, Mori M, Wada-Hiraike O, Oda K, Suzuki H, Maruyama R, and Osuga Y
- Subjects
- Male, Female, Humans, Nuclear Proteins genetics, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases metabolism, Histone Code, Apoptosis, Interferons, Histones metabolism, Endometrial Neoplasms genetics
- Abstract
Histone modification, a major epigenetic mechanism regulating gene expression through chromatin remodeling, introduces dynamic changes in chromatin architecture. Protein arginine methyltransferase 6 (PRMT6) is overexpressed in various types of cancer, including prostate, lung and endometrial cancer (EC). Epigenome regulates the expression of endogenous retrovirus (ERV), which activates interferon signaling related to cancer. The antitumor effects of PRMT6 inhibition and the role of PRMT6 in EC were investigated, using epigenome multi‑omics analysis, including an assay for chromatin immunoprecipitation sequencing (ChIP‑seq) and RNA sequencing (RNA‑seq). The expression of PRMT6 in EC was analyzed using reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and immunohistochemistry (IHC). The prognostic impact of PRMT6 expression was evaluated using IHC. The effects of PRMT6‑knockdown (KD) were investigated using cell viability and apoptosis assays, as well as its effects on the epigenome, using ChIP‑seq of H3K27ac antibodies and RNA‑seq. Finally, the downstream targets identified by multi‑omics analysis were evaluated. PRMT6 was overexpressed in EC and associated with a poor prognosis. PRMT6‑KD induced histone hypomethylation, while suppressing cell growth and apoptosis. ChIP‑seq revealed that PRMT6 regulated genomic regions related to interferons and apoptosis through histone modifications. The RNA‑seq data demonstrated altered interferon‑related pathways and increased expression of tumor suppressor genes, including NK6 homeobox 1 and phosphoinositide‑3‑kinase regulatory subunit 1, following PRMT6‑KD. RT‑qPCR revealed that eight ERV genes which activated interferon signaling were upregulated by PRMT6‑KD. The data of the present study suggested that PRMT6 inhibition induced apoptosis through interferon signaling activated by ERV. PRMT6 regulated tumor suppressor genes and may be a novel therapeutic target, to the best of our knowledge, in EC.
- Published
- 2024
- Full Text
- View/download PDF
25. Design, synthesis and evaluation of antitumor activity of selective PRMT6 inhibitors.
- Author
-
Zhang, Qiangsheng, Cao, Jiaying, Zhang, Yiqian, Bi, Zhenfei, Feng, Qiang, Yu, Luoting, and Li, Lu
- Subjects
- *
ANTINEOPLASTIC agents , *PROTEIN arginine methyltransferases , *HYDROGEN bonding interactions , *CANCER cells , *BINDING sites - Abstract
PRMT6 is a member of the protein arginine methyltransferase family, which is involved in a variety of physiological processes and plays an important role in the occurrence and development of tumors. Due to the high homology of type Ⅰ PRMTs and the two close binding sites of the SAM pocket and the substrate pocket, selective PRMT6 inhibitors have rarely been reported. In this study, a series of (5-phenylpyridin-3-yl)methanamine derivatives were designed and synthesized, which could form hydrogen bonding interactions with the unique Glu49 of PRMT6, thereby improving the selectivity of the compounds for PRMT6. Among them, a25 had the best activity and selectivity, with more than 25-fold selectivity for PRMT1/8 and more than 50-fold selectivity for PRMT3/4/5/7, which was superior to these reported SAM competitive and substrate competitive PRMT6 inhibitors. Importantly, a25 could significantly inhibit the proliferation of various tumor cells and effectively induce apoptosis of cancer cells. Our data clarified that a25 is a promising selective PRMT6 inhibitor for cancer therapy which is worthy of further evaluation. [Display omitted] • A series of novel and selective PRMT6 inhibitors have been synthesized. • a25 was 30-fold selective for PRMT1 and 28-fold selective for PRMT8. • a25 significantly inhibit the proliferation of various tumor cells. • a25 showed reasonable pharmacokinetic profile. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. PRMT6 inhibits K63-linked ubiquitination and promotes the degradation of IRF3 in the antiviral innate immunity of black carp Mylopharyngodon piceus.
- Author
-
Yang, Can, Yang, Shisi, Miao, Yujia, Shu, Juanjuan, Peng, Yuqing, Li, Jun, Wu, Hui, Zou, Jun, and Feng, Hao
- Subjects
- *
PROTEIN arginine methyltransferases , *CARP , *UBIQUITINATION , *NATURAL immunity , *CTENOPHARYNGODON idella , *INTERFERONS - Abstract
Protein arginine methyltransferases (PRMT6) has been shown to negatively regulate interferon (IFN) production in mammals, however, the role of teleost PRMT6 in IFN signaling remains largely unknown. In our previous study, black carp (Mylopharyngodon piceus) PRMT6 (bcPRMT6) has been identified to inhibit bcTBK1/bcIRF3 cascade. In this study, bcIRF3-induced transcription of bcIFNa, bcIFNb, bcp50 and bcIl1β in host cells was attenuated by co-expressed bcPRMT6. Accordingly, bcIRF3-mediated antiviral activity against grass carp reovirus (GCRV) and spring viremia of carp virus (SVCV) was obviously dampened by bcPRMT6. Knockdown of bcPRMT6 enhanced the antiviral ability of host cells and improved the transcription of bcIFNa, bcViperin and bcIl1β. When co-expressed with bcPRMT6 in either mammalian or fish cells, the protein level of bcIRF3 was decreased, which implied the triggered degradation of bcIRF3 by bcPRMT6. And interestingly, this decreased protein expression of bcIRF3 was rescued by chloroquine but not MG132, indicating the lysosome-dependent degradation of this molecule. The subsequent immunoprecipitation and immunoblot assay demonstrated that K63-linked ubiquitination but not K48-linked of bcIRF3 was obviously attenuated by bcPRMT6. And the immunofluorescent staining data suggested that the nuclear translocation of bcIRF3 was inhibited by bcPRMT6. Taken together, our data concludes that bcPRMT6 promotes the degradation of bcIRF3 through lysosome pathway and inhibits K63-linked ubiquitination of bcIRF3, leading to the dampened antiviral signaling mediated by bcIRF3. • Black carp PRMT6 promotes lysosome-dependent degradation of black carp IRF3. • Back carp PRMT6 dampens IRF3-mediated antiviral signaling. • Black carp PRMT6 inhibits K63-linked ubiquitination of black carp IRF3. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. PRMT6 functionally associates with PRMT5 to promote colorectal cancer progression through epigenetically repressing the expression of CDKN2B and CCNG1.
- Author
-
Chen, Yuzhong, Liang, Wanqing, Du, Jun, Ma, Jiachi, Liang, Rongrui, and Tao, Min
- Subjects
- *
COLORECTAL cancer , *PROTEIN arginine methyltransferases , *CANCER invasiveness , *GENETIC regulation , *MASS spectrometry , *IMMUNOPRECIPITATION - Abstract
Protein arginine methyltransferase 6 (PRMT6) is a type I arginine methyltransferase that asymmetrically dimethylates histone H3 arginine 2 (H3R2me2a). However, the biological roles and underlying molecular mechanisms of PRMT6 in colorectal cancer (CRC) remain unclear. PRMT6 expression in CRC tissue was examined using immunohistochemistry. The effect of PRMT6 on CRC cells was investigated in vitro and in vivo. Mass spectrometry, co-immunoprecipitation and GST pulldown assays were performed to identify interaction partners of PRMT6. RNA-seq, chromatin immunoprecipitation, Western blot and qRT-PCR assays were used to investigate the mechanism of PRMT6 in gene regulation. PRMT6 is significantly upregulated in CRC tissues and facilitates cell proliferation of CRC cells in vitro and in vivo. Through RNA-seq analysis, CDKN2B (p15INK4b) and CCNG1 were identified as new transcriptional targets of PRMT6. PRMT6-dependent H3R2me2a mark was predominantly deposited at the promoters of CDKN2B and CCNG1 in CRC cells. Furthermore, PRMT5 was firstly characterized as an interaction partner of PRMT6. Notably, H3R2me2a coincides with PRMT5-mediated H4R3me2s and H3R8me2s marks at the promoters of CDKN2B and CCNG1 genes, thus leading to transcriptional repression of these genes. PRMT6 functionally associates with PRMT5 to promote CRC progression through epigenetically repressing the expression of CDKN2B and CCNG1. These insights raise the possibility that combinational intervention of PRMT6 and PRMT5 may be a promising strategy for CRC therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Decoding m 6 A RNA methylome identifies PRMT6-regulated lipid transport promoting AML stem cell maintenance.
- Author
-
Cheng Y, Gao Z, Zhang T, Wang Y, Xie X, Han G, Li Y, Yin R, Chen Y, Wang P, Hu J, Zhang T, Guo C, Chai J, Wang J, Cui M, Gao K, Liu W, Yao S, Lu P, Cao Z, Zheng Y, Chang J, Liu Z, Song Q, Li W, Zhou F, and Zhang H
- Subjects
- Humans, Animals, Mice, Epigenome, RNA, Messenger metabolism, Neoplastic Stem Cells metabolism, Lipids, Mammals metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Nuclear Proteins metabolism, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases metabolism, RNA metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism
- Abstract
N
6 -methyladenosine (m6 A) is a common chemical modification for mammalian mRNA and exhibits high dynamics in various biological processes. However, dynamics of m6 A RNA methylome during leukemogenesis remains unknown. Here, we delineate a comprehensive m6 A landscape during acute myeloid leukemia (AML) development and identify PRMT6 as a key for maintaining AML stem cells. We observe an obvious change in m6 A methylome during leukemogenesis and find that protein arginine methyltransferase PRMT6 and m6 A reader IGF2BP2 maintain the function of human and murine leukemia stem cells (LSCs). Genetic deletion or pharmacological inhibition of PRMT6 damages AML development and LSC function. Mechanistically, IGF2BP2 stabilizes PRMT6 mRNA via m6 A-mediated manner, which catalyzes H3R2me2a and suppresses lipid transporter MFSD2A expression. PRMT6 loss upregulates MFSD2A expression that increases docosahexaenoic acid levels and impairs LSC maintenance. Collectively, our findings reveal a critical role of PRMT6-MFSD2A signaling axis in AML development and provide a therapeutic strategy for targeting LSCs., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
29. Grass carp PRMT6 negatively regulates innate immunity by inhibiting the TBK1/IRF3 binding and cutting down IRF3 phosphorylation level.
- Author
-
Jiang, Zeyin, Cheng, Xining, Sun, Zhichao, Hu, Jihuan, Xu, Xiaowen, Li, Meifeng, Feng, Zhiqing, and Hu, Chengyu
- Subjects
- *
CTENOPHARYNGODON idella , *PROTEIN arginine methyltransferases , *METHYLTRANSFERASES , *PHOSPHORYLATION , *HISTONES , *AMINO acids - Abstract
Subcellular localization analysis implicated that CiPRMT6 was mainly located in the nucleus, with a small part of them located in the cytoplasm. PRMT6, namely protein arginine methyltransferase 6, was first identified and demonstrated to catalyze the methylation of arginine residue on the chromatin histones in mammals. Mammalian PRMT6 usually acts as an arginine methyltransferase in the nucleus, but induces antiviral innate immune response in the cytoplasm. Nowadays, there have been few reports about PRMT6 in teleost. In this study, we investigated the potential molecular mechanisms underlying the interaction of PRMT6 expression and IFN1 response in grass carp. We first cloned and identified a grass carp PRMT6 (named CiPRMT6, MN781672.1), which is 1068bp in length encoding a deduced polypeptide of 355 amino acids. In CIK cell, CiPRMT6 expression was up-regulated upon stimulation with poly (I:C); while overexpression of PRMT6 suppressed the promoter activity of grass carp IFN1 and reduced the phosphorylation of IRF3; however, the amount of PRMT6 mutant (lack of methyltransferase domain) was increased in the cytoplasm. Our results also showed that grass carp PRMT6 and IRF3 (but not TBK1) were co-located and bound to each other in the cytoplasm. The binding of CiPRMT6 to IRF3 impairs the interaction between TBK1 and IRF3, indicating that CiPRMT6 is a negative regulator for IFN1 expression through TBK1-IRF3 signaling pathway in grass carp. In conclusion, we identified that CiPRMT6 negatively regulated IFN1 expression by inhibiting the TBK1-IRF3 interaction as well as IRF3 phosphorylation. • Grass carp PRMT6 can be upregulated by poly I:C. • Grass carp PRMT6 reduces the phosphorylation level of IRF3. • The binding of grass carp PRMT6 to IRF3 impairs grass carp TBK1/IRF3 binding. • Grass carp PRMT6 reduces IFN1 expression by blocking TBK1-IRF3. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.