882 results on '"PRMT5"'
Search Results
2. Shedding light on imaging cancer research: Design and synthesis of 1, 8-naphthalimide-based PRMT5-targeted fluorescent ligands
- Author
-
Ali Shah, Sayed Asmat, Guo, Zihao, Zhang, Peng, Bian, Shaopan, Ma, Yanan, Li, Shufeng, Wang, Xiaodi, Wu, Di, Zhang, Hang, and Xu, Haiwei
- Published
- 2025
- Full Text
- View/download PDF
3. First-in-human study of AMG 193, an MTA-cooperative PRMT5 inhibitor, in patients with MTAP-deleted solid tumors: results from phase I dose exploration
- Author
-
Rodon, J., Prenen, H., Sacher, A., Villalona-Calero, M., Penel, N., El Helali, A., Rottey, S., Yamamoto, N., Ghiringhelli, F., Goebeler, M.E., Doi, T., Postel-Vinay, S., Lin, C.-C., Liu, C., Chuang, C.-H., Keyvanjah, K., Eggert, T., and O’Neil, B.H.
- Published
- 2024
- Full Text
- View/download PDF
4. Research progress in DNA damage response (DDR)-targeting modulators: From hits to clinical candidates
- Author
-
Cheng, Binbin, Ding, Zongbao, Hong, Yimeng, Wang, Yaping, Zhou, Yingxing, Chen, Jianjun, Peng, Xiaopeng, and Zeng, Chunlai
- Published
- 2025
- Full Text
- View/download PDF
5. Nutrient control of splice site selection contributes to methionine addiction of cancer
- Author
-
Lin, Da-Wei, Carranza, Francisco G., Borrego, Stacey, Lauinger, Linda, Dantas de Paula, Lucas, Pulipelli, Harika R., Andronicos, Anna, Hertel, Klemens J., and Kaiser, Peter
- Published
- 2025
- Full Text
- View/download PDF
6. Structure-based discovery of a new series of nucleoside-derived ring-opening PRMT5 inhibitors
- Author
-
Chen, Yuting, Wang, Zekun, Zhang, Junjie, Shi, Qiongyu, Yang, Hong, Deng, Yue, Wang, Xingcan, Liu, Tongchao, Geng, Meiyu, Xiong, Bing, and Huang, Xun
- Published
- 2024
- Full Text
- View/download PDF
7. Tumor-intrinsic PRMT5 upregulates FGL1 via methylating TCF12 to inhibit CD8+ T-cell-mediated antitumor immunity in liver cancer
- Author
-
Sun, Jiao, Yuan, Hongfeng, Sun, Linlin, Zhao, Lina, Wang, Yufei, Hou, Chunyu, Zhang, Huihui, Lv, Pan, Yang, Guang, Zhang, Ningning, Lu, Wei, and Zhang, Xiaodong
- Published
- 2024
- Full Text
- View/download PDF
8. PRMT5 knockdown enhances cell viability and suppresses cell apoptosis, oxidative stress, inflammation and endothelial dysfunction in ox-LDL-induced vascular endothelial cells via interacting with PDCD4
- Author
-
Fei, Xiaohong, Cen, Xuejiang, Zhao, Ruochi, Wang, Jian, and Cui, Hanbin
- Published
- 2023
- Full Text
- View/download PDF
9. Role of PRMT5 mediated HOXA10 arginine 337 methylation in endometrial epithelial cell receptivity
- Author
-
Cao, Zhiwen, Jiang, Jinwen, Wang, Yiting, Lu, Yuhang, Wu, Min, Zhen, Xin, Cai, Xinyu, Sun, Haixiang, and Yan, Guijun
- Published
- 2024
- Full Text
- View/download PDF
10. Prmt5 is essential for intestinal stem cell maintenance and homeostasis.
- Author
-
Yang, Li, Li, Xuewen, Shi, Chenyi, and Zhao, Bing
- Abstract
Intestinal homeostasis relies on the continuous renewal of intestinal stem cells (ISCs), which could be epigenetically regulated. While protein arginine methyltransferase 5 (Prmt5) is known to play a key role in multiple organs as an epigenetic modifier, its specific function in maintaining intestinal homeostasis remains to be elucidated. Here, we show that Prmt5 is highly expressed in mouse crypts. The deletion of Prmt5 results in ISCs deficiency, ectopic localization of Paneth cells, and spontaneous colitis. Mechanistically, Prmt5 sustains a high level of H3K27ac accumulation by inhibiting Hdac9 expression in the intestinal epithelium, and maintains the stemness of ISCs in a cell-autonomous manner. Notably, inhibition of histone deacetylases can rescue both self‐renewal and differentiation capacities of Prmt5‐depleted ISCs. These findings highlight Prmt5 as a critical regulator in intestinal epithelium development and tissue homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. PRMT5 Inhibitor EPZ015666 Decreases the Viability and Encystment of Entamoeba invadens.
- Author
-
Ortiz-Hernández, Rigoberto, Millán-Casarrubias, Elmer Joel, Bolaños, Jeni, Munguía-Robledo, Susana, Vázquez-Calzada, Carlos, Azuara-Licéaga, Elisa, Valdés, Jesús, and Rodríguez, Mario Alberto
- Subjects
- *
BINDING sites , *PROTEIN arginine methyltransferases , *ENTAMOEBA histolytica , *LIFE cycles (Biology) , *ENCYSTMENT - Abstract
Protein arginine methyltransferase 5 (PRMT5) is an enzyme that produces monomethyl arginine (MMA) and symmetric dimethyl arginine (sDMA), post-translational modifications that regulate several cellular processes, including stage conversion in parasitic protozoans. Entamoeba histolytica, the etiologic agent of human amebiasis, has two stages in its life cycle, the trophozoite, which is the replicative form, and the cyst, corresponding to the infective phase. The study of the molecular mechanisms that regulate differentiation in this parasite has been overdue because of a lack of efficient protocols for in vitro encystment. For this reason, Entamoeba invadens, a parasite of reptiles, has been used as a differentiation model system for the genus. Here, we demonstrated the presence of sDMA in E. invadens, which increases during encystment, and identified the PRMT5 of this microorganism (EiPRMT5). In addition, we performed 3D modeling of this enzyme, as well as its molecular docking with the PRMT5 inhibitor EPZ015666, which predicted the affinity of the drug for the active site of the enzyme. In agreement with these findings, EPZ015666 reduced trophozoite viability and encystment. Therefore, EiPRMT5 is a potential target for inhibiting the spread of amebiasis. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
12. Tumor-intrinsic PRMT5 upregulates FGL1 via methylating TCF12 to inhibit CD8+ T-cell-mediated antitumor immunity in liver cancer.
- Author
-
Sun, Jiao, Yuan, Hongfeng, Sun, Linlin, Zhao, Lina, Wang, Yufei, Hou, Chunyu, Zhang, Huihui, Lv, Pan, Yang, Guang, Zhang, Ningning, Lu, Wei, and Zhang, Xiaodong
- Subjects
IMMUNE checkpoint proteins ,PROTEIN arginine methyltransferases ,LIVER cancer ,IMMUNOSUPPRESSION ,TREATMENT effectiveness - Abstract
Protein arginine methyltransferase 5 (PRMT5) acts as an oncogene in liver cancer, yet its roles and in-depth molecular mechanisms within the liver cancer immune microenvironment remain mostly undefined. Here, we demonstrated that disruption of tumor-intrinsic PRMT5 enhances CD8
+ T-cell-mediated antitumor immunity both in vivo and in vitro. Further experiments verified that this effect is achieved through downregulation of the inhibitory immune checkpoint molecule, fibrinogen-like protein 1 (FGL1). Mechanistically, PRMT5 catalyzed symmetric dimethylation of transcription factor 12 (TCF12) at arginine 554 (R554), prompting the binding of TCF12 to FGL1 promoter region, which transcriptionally activated FGL1 in tumor cells. Methylation deficiency at TCF12-R554 residue downregulated FGL1 expression, which promoted CD8+ T-cell-mediated antitumor immunity. Notably, combining the PRMT5 methyltransferase inhibitor GSK591 with PD-L1 blockade efficiently inhibited liver cancer growth and improved overall survival in mice. Collectively, our findings reveal the immunosuppressive role and mechanism of PRMT5 in liver cancer and highlight that targeting PRMT5 could boost checkpoint immunotherapy efficacy. Tumor-intrinsic PRMT5 upregulates FGL1 via methylating TCF12 to inhibit CD8+ T-cell-mediated antitumor immunity in liver cancer. Combining the PRMT5 methyltransferase inhibitor GSK591 with PD-L1 blockade results in augmented antitumor immunity. [Display omitted] [ABSTRACT FROM AUTHOR]- Published
- 2025
- Full Text
- View/download PDF
13. A comprehensive investigation of PRMT5 in the prognosis and ion channel features of lung cancer.
- Author
-
Wang, Yan, Chu, Daifang, Li, Haichao, Fan, Jiangjiang, Zhu, Ximing, Ma, Yulong, Gu, Zhongping, Xie, Nianlin, and Jing, Pengyu
- Subjects
PROGRESSION-free survival ,ALTERNATIVE RNA splicing ,SURVIVAL rate ,PROTEIN arginine methyltransferases ,OVERALL survival - Abstract
The increasing incidence and mortality associated with lung cancer (LC) is a significant global health challenge. The underlying mechanisms contributing to LC remain inadequately understood. However, emerging evidence suggests that the epigenetic modifier protein arginine methyltransferase 5 (PRMT5) plays a complex role in various cellular processes, including DNA repair, gene transcription, and alternative splicing, through its function in catalyzing the symmetric dimethylation of both histone and non-histone proteins. In this study, we examined the functional role of PRMT5 utilizing LC-related datasets (GSE30219, GSE50081, and TCGA LC cohort) through a series of analyses. Our findings revealed that PRMT5 was significantly overexpressed in LC samples compared to normal tissues and was correlated with overall survival and disease-free survival rates. Additionally, PRDM1 was identified as a key protein exhibiting a strong interaction with PRMT5. The prognostic model that integrated PRMT5 with clinical factors demonstrated robust performance in assessing survival outcomes. Elevated levels of PRMT5 were associated with poor prognosis in LC, as evidenced by analyses of the GSE30219, GSE50081, and TCGA-LC datasets. Furthermore, we identified 27 ion channel (IC) genes exhibited a correlation with PRMT5 in lung adenocarcinoma (LUAD), of which 9 genes were identified as statistically significant with KM survival analysis. Strikingly, all of the 9 genes, including LRRC8A, the same as PRMT5, were associated with poor prognosis in LUAD. Our research highlights the potential of PRMT5 as a novel prognostic biomarker and its relationship with IC genes in LC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Application and research progress of synthetic lethality in the development of anticancer therapeutic drugs.
- Author
-
Gong, Xiaoliang, Liu, Chunxi, Tang, Haoyang, Wu, Song, and Yang, Qingyun
- Subjects
POLY(ADP-ribose) polymerase ,PROTEIN arginine methyltransferases ,WERNER'S syndrome ,ANTINEOPLASTIC agents ,DRUG development - Abstract
With the tremendous success of the PARP inhibitor olaparib in clinical practice, synthetic lethality has become an important field for the discovery and development of anticancer drugs. More and more synthetic lethality targets have been discovered with the rapid development of biotechnology in recent years. Currently, many drug candidates that were designed and developed on the basis of the concept of synthetic lethality have entered clinical trials. Taking representative synthetic lethal targets Poly ADP-ribose polymerase 1 (PARP1), Werner syndrome helicase (WRN) and protein arginine methyltransferase 5 (PRMT5) as examples, this article briefly discusses the application and research progress of synthetic lethality in the development of anticancer drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Prmt5 is essential for intestinal stem cell maintenance and homeostasis
- Author
-
Li Yang, Xuewen Li, Chenyi Shi, and Bing Zhao
- Subjects
Prmt5 ,Intestinal stem cells ,Homeostasis ,H3K27ac ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Abstract Intestinal homeostasis relies on the continuous renewal of intestinal stem cells (ISCs), which could be epigenetically regulated. While protein arginine methyltransferase 5 (Prmt5) is known to play a key role in multiple organs as an epigenetic modifier, its specific function in maintaining intestinal homeostasis remains to be elucidated. Here, we show that Prmt5 is highly expressed in mouse crypts. The deletion of Prmt5 results in ISCs deficiency, ectopic localization of Paneth cells, and spontaneous colitis. Mechanistically, Prmt5 sustains a high level of H3K27ac accumulation by inhibiting Hdac9 expression in the intestinal epithelium, and maintains the stemness of ISCs in a cell-autonomous manner. Notably, inhibition of histone deacetylases can rescue both self‐renewal and differentiation capacities of Prmt5‐depleted ISCs. These findings highlight Prmt5 as a critical regulator in intestinal epithelium development and tissue homeostasis.
- Published
- 2025
- Full Text
- View/download PDF
16. Targeting PRMT5 through PROTAC for the treatment of triple-negative breast cancer
- Author
-
Yaxun Guo, Yuzhan Li, Zhongmei Zhou, Lei Hou, Wenjing Liu, Wenlong Ren, Dazhao Mi, Jian Sun, Xueqin Dai, Yingying Wu, Zhuo Cheng, Tingyue Wu, Qianmei Luo, Cong Tian, Fubing Li, Zhigang Yu, Yihua Chen, and Ceshi Chen
- Subjects
PROTAC ,PRMT5 ,KLF5 ,TNBC ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Triple-negative breast cancer (TNBC) is currently the most aggressive subtype of breast cancer, characterized by high heterogeneity and strong invasiveness, and currently lacks effective therapies. PRMT5, a type II protein arginine methyltransferase, is upregulated in numerous cancers, including TNBC, and plays a critical role, marked it as an attractive therapeutic target. PROTAC (Proteolysis Targeting Chimeras) is an innovative drug development technology that utilizes the ubiquitin-proteasome system (UPS) to degrade target proteins, which is characterized by higher activity, enhanced safety, lower resistance, and reduced toxicity, offering significant value for clinical translation. Methods This study utilizes the PROTAC technology to develop potential degraders targeting PRMT5 in vitro and in vivo. Results Through the design, synthesis and screening of a series of targeted compounds, we identified YZ-836P as an effective compound that exerted cytotoxic effects and reduced the protein levels of PRMT5 and its key downstream target protein KLF5 in TNBC after 48 h. Its efficacy was significantly superior to the PRMT5 PROTAC degraders that had been reported. YZ-836P induced G1 phase cell cycle arrest and significantly induced apoptosis in TNBC cells. Additionally, we demonstrated that YZ-836P promoted the ubiquitination and degradation of PRMT5 in a cereblon (CRBN)-dependent manner. Notably, YZ-836P exhibited pronounced efficacy in inhibiting the growth of TNBC patient-derived organoids and xenografts in nude mice. Conclusions These findings position YZ-836P as a promising candidate for advancing treatment modalities for TNBC. Trial registration Ethics Committee of Yunnan Cancer Hospital, KYCS2023-078. Registered 7 June 2023.
- Published
- 2024
- Full Text
- View/download PDF
17. Targeting PRMT5 through PROTAC for the treatment of triple-negative breast cancer.
- Author
-
Guo, Yaxun, Li, Yuzhan, Zhou, Zhongmei, Hou, Lei, Liu, Wenjing, Ren, Wenlong, Mi, Dazhao, Sun, Jian, Dai, Xueqin, Wu, Yingying, Cheng, Zhuo, Wu, Tingyue, Luo, Qianmei, Tian, Cong, Li, Fubing, Yu, Zhigang, Chen, Yihua, and Chen, Ceshi
- Subjects
TRIPLE-negative breast cancer ,PROTEIN arginine methyltransferases ,BREAST cancer ,GENETIC translation ,CANCER hospitals - Abstract
Background: Triple-negative breast cancer (TNBC) is currently the most aggressive subtype of breast cancer, characterized by high heterogeneity and strong invasiveness, and currently lacks effective therapies. PRMT5, a type II protein arginine methyltransferase, is upregulated in numerous cancers, including TNBC, and plays a critical role, marked it as an attractive therapeutic target. PROTAC (Proteolysis Targeting Chimeras) is an innovative drug development technology that utilizes the ubiquitin-proteasome system (UPS) to degrade target proteins, which is characterized by higher activity, enhanced safety, lower resistance, and reduced toxicity, offering significant value for clinical translation. Methods: This study utilizes the PROTAC technology to develop potential degraders targeting PRMT5 in vitro and in vivo. Results: Through the design, synthesis and screening of a series of targeted compounds, we identified YZ-836P as an effective compound that exerted cytotoxic effects and reduced the protein levels of PRMT5 and its key downstream target protein KLF5 in TNBC after 48 h. Its efficacy was significantly superior to the PRMT5 PROTAC degraders that had been reported. YZ-836P induced G1 phase cell cycle arrest and significantly induced apoptosis in TNBC cells. Additionally, we demonstrated that YZ-836P promoted the ubiquitination and degradation of PRMT5 in a cereblon (CRBN)-dependent manner. Notably, YZ-836P exhibited pronounced efficacy in inhibiting the growth of TNBC patient-derived organoids and xenografts in nude mice. Conclusions: These findings position YZ-836P as a promising candidate for advancing treatment modalities for TNBC. Trial registration: Ethics Committee of Yunnan Cancer Hospital, KYCS2023-078. Registered 7 June 2023. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. A chemical screen identifies PRMT5 as a therapeutic vulnerability for paclitaxel-resistant triple-negative breast cancer.
- Author
-
Zhang, KeJing, Wei, Juan, Zhang, SheYu, Fei, Liyan, Guo, Lu, Liu, Xueying, Ji, YiShuai, Chen, WenJun, Ciamponi, Felipe E., Chen, WeiChang, Li, MengXi, Zhai, Jie, Fu, Ting, Massirer, Katlin B., Yu, Yang, Lupien, Mathieu, Wei, Yong, Arrowsmith, Cheryl. H., Wu, Qin, and Tan, WeiHong
- Subjects
- *
PROTEIN arginine methyltransferases , *TRIPLE-negative breast cancer , *AURORA kinases , *PACLITAXEL , *BREAST cancer , *PROTEIN expression , *RNA splicing - Abstract
Paclitaxel-resistant triple negative breast cancer (TNBC) remains one of the most challenging breast cancers to treat. Here, using an epigenetic chemical probe screen, we uncover an acquired vulnerability of paclitaxel-resistant TNBC cells to protein arginine methyltransferases (PRMTs) inhibition. Analysis of cell lines and in-house clinical samples demonstrates that resistant cells evade paclitaxel killing through stabilizing mitotic chromatin assembly. Genetic or pharmacologic inhibition of PRMT5 alters RNA splicing, particularly intron retention of aurora kinases B (AURKB), leading to a decrease in protein expression, and finally results in selective mitosis catastrophe in paclitaxel-resistant cells. In addition, type I PRMT inhibition synergies with PRMT5 inhibition in suppressing tumor growth of drug-resistant cells through augmenting perturbation of AURKB-mediated mitotic signaling pathway. These findings are fully recapitulated in a patient-derived xenograft (PDX) model generated from a paclitaxel-resistant TNBC patient, providing the rationale for targeting PRMTs in paclitaxel-resistant TNBC. [Display omitted] • Paclitaxel-resistant triple-negative breast cancer is vulnerable to PRMTs inhibition • Inhibiting type I PRMTs and PRMT5 synergizes to suppress paclitaxel-resistant TNBC growth • Targeting PRMTs induces intron retention in AURKB, leading to mitotic catastrophe Paclitaxel-resistant triple-negative breast cancer (TNBC) remains challenging to treat. Zhang et al. discover that resistant TNBC cells are vulnerable to PRMT inhibition, which decreases AURKB protein levels and induces mitotic catastrophe. Inhibiting type I PRMTs and PRMT5 synergizes to suppress tumor growth, suggesting a promising strategy for treating paclitaxel-resistant TNBC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. PRMT5/WDR77 Enhances the Proliferation of Squamous Cell Carcinoma via the ΔNp63α-p21 Axis.
- Author
-
Liang, Heng, Fisher, Matthew L., Wu, Caizhi, Ballon, Carlos, Sun, Xueqin, and Mills, Alea A.
- Subjects
- *
SQUAMOUS cell carcinoma , *ARGININE , *FLOW cytometry , *PROTEIN kinase inhibitors , *GENOMICS , *RESEARCH funding , *HEAD & neck cancer , *CELL proliferation , *TREATMENT effectiveness , *DESCRIPTIVE statistics , *REVERSE transcriptase polymerase chain reaction , *METHYLTRANSFERASES , *CELL culture , *IMMUNOHISTOCHEMISTRY , *WESTERN immunoblotting , *GENE expression profiling , *DATA analysis software , *SEQUENCE analysis - Abstract
Simple Summary: Protein arginine methyltransferase 5 (PRMT5) is known to be oncogenic in many cancers, including squamous cell carcinoma (SCC). Our analyses of multiple public databases revealed that PRMT5 overexpression correlates with poor survival in SCC patients and is essential to the survival of SCC cell lines. This study focused on understanding how PRMT5 and its binding partner, WDR77 (WD repeat domain 77), regulate SCC cell growth, particularly through the p63 ΔNp63α isoform, a key factor in SCC. Furthermore, PRMT5 depletion inhibited SCC proliferation by inducing cell cycle arrest in the G1 phase. Additionally, we showed that PRMT5 and WDR77 stabilized ΔNp63α protein expression, which in turn inhibited p21 (cyclin-dependent kinase inhibitor 1). These findings provide new insights into the potential of targeting PRMT5 as a therapeutic strategy for SCC. Protein arginine methyltransferase 5 (PRMT5) is a critical oncogenic factor in various cancers, and its inhibition has shown promise in suppressing tumor growth. However, the role of PRMT5 in squamous cell carcinoma (SCC) remains largely unexplored. In this study, we analyzed SCC patient data from The Cancer Genome Atlas (TCGA) and the Cancer Dependency Map (DepMap) to investigate the relationship between PRMT5 and SCC proliferation. We employed competition-based cell proliferation assays, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays, flow cytometry, and in vivo mouse modeling to examine the regulatory roles of PRMT5 and its binding partner WDR77 (WD repeat domain 77). We identified downstream targets, including the p63 isoform ΔNp63α and the cyclin-dependent kinase inhibitor p21, through single-cell RNA-seq, RT-qPCR, and Western blot analyses. Our findings demonstrate that upregulation of PRMT5 and WDR77 correlates with the poor survival of head and neck squamous cell carcinoma (HNSCC) patients. PRMT5/WDR77 regulates the HNSCC-specific transcriptome and facilitates SCC proliferation by promoting cell cycle progression. The PRMT5 and WDR77 stabilize the ΔNp63α Protein, which in turn, inhibits p21. Moreover, depletion of PRMT5 and WDR77 repress SCC in vivo. This study reveals for the first time that PRMT5 and WDR77 synergize to promote SCC proliferation via the ΔNp63α-p21 axis, highlighting a novel therapeutic target for SCC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. SCR‐7952, a highly selective MAT2A inhibitor, demonstrates synergistic antitumor activities in combination with the S‐adenosylmethionine‐competitive or the methylthioadenosine‐cooperative protein arginine methyltransferase 5 inhibitors in methylthioadenosine phosphorylase‐deleted tumors
- Author
-
Yu, Zhiyong, Kuang, Yi, Xue, Liting, Ma, Xuan, Li, Tingting, Yuan, Linlin, Li, Mengying, Xue, Grace, Li, Zhen, Tang, Feng, Tang, Jianxing, Shan, Jinwen, Wang, Weijie, Tang, Renhong, and Zhou, Feng
- Subjects
PROTEIN arginine methyltransferases ,BINDING sites ,ENZYMES ,ANTINEOPLASTIC agents ,BILIRUBIN - Abstract
The metabolic enzyme methionine adenosyltransferase 2A (MAT2A) was found to elicit synthetic lethality in methylthioadenosine phosphorylase (MTAP)‐deleted cancers, which occur in about 15% of all cancers. Here, we described a novel MAT2A inhibitor, SCR‐7952 with potent and selective antitumor effects on MTAP‐deleted cancers in both in vitro and in vivo. The cryo‐EM data indicated the high binding affinity and the allosteric binding site of SCR‐7952 on MAT2A. Different from AG‐270, SCR‐7952 exhibited little influence on metabolic enzymes and did not increase the plasma levels of bilirubin. A systematic evaluation of combination between SCR‐7952 and different types of protein arginine methyltransferase 5 (PRMT5) inhibitors indicated remarkable synergistic interactions between SCR‐7952 and the S‐adenosylmethionine‐competitive or the methylthioadenosine‐cooperative PRMT5 inhibitors, but not substrate‐competitive ones. The mechanism was via the aggravated inhibition of PRMT5 and FANCA splicing perturbations. These results indicated that SCR‐7952 could be a potential therapeutic candidate for the treatment of MTAP‐deleted cancers, both monotherapy and in combination with PRMT5 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. The crescent-like Golgi ribbon is shaped by the Ajuba/PRMT5/Aurora-A complex-modified HURP.
- Author
-
Chiu, Shao-Chih, Yang, Xin-Ting, Wei, Tong-You, Liao, Yu-Ting, Chen, Jo-Mei, Kuo, Yi-Chun, Liu, Chun-Chih, Cheng, Chiao-Yun, Huang, Yu-Ting, Huang, Yun-Ru, Wu, He-Lian, Wan, Chang-Xin, Tsai, Jia-Rung, and Yu, Chang-Tze
- Subjects
ARF1 ,Ajuba ,Aurora-A ,Golgi ribbon ,HURP ,PRMT5 ,Animals ,Humans ,Golgi Apparatus ,Phosphorylation ,Cell Nucleus ,Protein-Arginine N-Methyltransferases ,Mammals - Abstract
BACKGROUND: Golgi apparatus (GA) is assembled as a crescent-like ribbon in mammalian cells under immunofluorescence microscope without knowing the shaping mechanisms. It is estimated that roughly 1/5 of the genes encoding kinases or phosphatases in human genome participate in the assembly of Golgi ribbon, reflecting protein modifications play major roles in building Golgi ribbon. METHODS: To explore how Golgi ribbon is shaped as a crescent-like structure under the guidance of protein modifications, we identified a protein complex containing the scaffold proteins Ajuba, two known GA regulators including the protein kinase Aurora-A and the protein arginine methyltransferase PRMT5, and the common substrate of Aurora-A and PRMT5, HURP. Mutual modifications and activation of PRMT5 and Aurora-A in the complex leads to methylation and in turn phosphorylation of HURP, thereby producing HURP p725. The HURP p725 localizes to GA vicinity and its distribution pattern looks like GA morphology. Correlation study of the HURP p725 statuses and GA structure, site-directed mutagenesis and knockdown-rescue experiments were employed to identify the modified HURP as a key regulator assembling GA as a crescent ribbon. RESULTS: The cells containing no or extended distribution of HURP p725 have dispersed GA membranes or longer GA. Knockdown of HURP fragmentized GA and HURP wild type could, while its phosphorylation deficiency mutant 725A could not, restore crescent Golgi ribbon in HURP depleted cells, collectively indicating a crescent GA-constructing activity of HURP p725. HURP p725 is transported, by GA membrane-associated ARF1, Dynein and its cargo adaptor Golgin-160, to cell center where HURP p725 forms crescent fibers, binds and stabilizes Golgi assembly factors (GAFs) including TRIP11, GRASP65 and GM130, thereby dictating the formation of crescent Golgi ribbon at nuclear periphery. CONCLUSIONS: The Ajuba/PRMT5/Aurora-A complex integrates the signals of protein methylation and phosphorylation to HURP, and the HURP p725 organizes GA by stabilizing and recruiting GAFs to its crescent-like structure, therefore shaping GA as a crescent ribbon. Therefore, the HURP p725 fiber serves a template to construct GA according to its shape. Video Abstract.
- Published
- 2023
22. Prognostic potential of PRMT5 and DSG2 proteins in pre-malignant cervical lesions
- Author
-
Bibiana Krajňáková, Desanka Výbohová, Sandra Hurta-Csizmár, Veronika Mešťanová, and Marian Adamkov
- Subjects
cervical lesions ,prmt5 ,dsg2 ,immunohistochemistry ,prognosis ,Medicine - Abstract
Precancerous cervical lesions are metaplastic alterations of epithelial cells of the cervix, eventually developing into cervical cancer. Despite primary and secondary prevention, the burden of cervical cancer remains high globally. Protein arginine methyltransferases (PRMT) represent post-translational modifications that interact with multiple signalling pathways, playing a role in epithelial-mesenchymal transition. In complex with desmoglein-2 (DSG2), a cell adhesion protein, both participate in the progression of dysplastic changes with potential malignant development. The presented study was performed on archival paraffin-embedded blocks from adult women. The studied samples were categorised into low-grade and high-grade intraepithelial lesions. Immunohistochemical analysis was used to observe subcellular localisation, immunoreaction intensity, and percentage of PRMT5- and DSG2-expressing cells, followed by statistical analysis. Preliminary results identified statistically significant differences between the expression and subcellular localisation of proteins in question in low-grade and high-grade squamous intraepithelial lesions. The primary goal of the presented study is to perceive the involvement of PRMT5 and DSG2 in the initiation and progression of cervical lesions. Our observations indicate the potential of the assessed proteins as prognostic markers. However, further studies of PRMT5 and DSG2 are required to provide greater insight into cervical carcinogenesis.
- Published
- 2024
- Full Text
- View/download PDF
23. Targeting PRMT5 enhances the radiosensitivity of tumor cells grown in vitro and in vivo
- Author
-
Charlotte Degorre, Steven Lohard, Christina N. Bobrek, Komal N. Rawal, Skyler Kuhn, and Philip J. Tofilon
- Subjects
PRMT5 ,Radiosensitization ,DNA damage response ,Medicine ,Science - Abstract
Abstract PRMT5 is a widely expressed arginine methyltransferase that regulates processes involved in tumor cell proliferation and survival. In the study described here, we investigated whether PRMT5 provides a target for tumor radiosensitization. Knockdown of PRMT5 using siRNA enhanced the radiosensitivity of a panel of cell lines corresponding to tumor types typically treated with radiotherapy. To extend these studies to an experimental therapeutic setting, the PRMT5 inhibitor LLY-283 was used. Exposure of the tumor cell lines to LLY-283 decreased PRMT5 activity and enhanced their radiosensitivity. This increase in radiosensitivity was accompanied by an inhibition of DNA double-strand break repair as determined by γH2AX foci and neutral comet analyses. For a normal fibroblast cell line, although LLY-283 reduced PRMT5 activity, it had no effect on their radiosensitivity. Transcriptome analysis of U251 cells showed that LLY-283 treatment reduced the expression of genes and altered the mRNA splicing pattern of genes involved in the DNA damage response. Subcutaneous xenografts were then used to evaluate the in vivo response to LLY-283 and radiation. Treatment of mice with LLY-283 decreased tumor PRMT5 activity and significantly enhanced the radiation-induced growth delay. These results suggest that PRMT5 is a tumor selective target for radiosensitization.
- Published
- 2024
- Full Text
- View/download PDF
24. Understanding the role of PRMT5 in neuroblastoma
- Author
-
Albayrak, Gulsah, La Thangue, Nicholas, and Humphrey, Timothy
- Subjects
PRMT5 ,Epigenetics ,Cancer ,Neuroblastoma - Abstract
Neuroblastoma (NB) is a deadly childhood cancer that has an orphan disease status, and the most malignant forms of the tumours have MYCN amplification. There is significant unmet medical need and urgency to develop new treatments for high-risk NB patients, as clinical research on rare diseases of children is very challenging. We revealed that higher PRMT5, MYCN, and E2F1 levels are poor prognostic factors in NB according to their Kaplan-Meier survival plots and then we hypothesized that PRMT5-E2F1-MYCN pathway might have potential prognostic value in NB. We characterized NB cell lines in terms of sensitivity to the PRMT5 inhibition by using genomic and transcriptomic assays in order to gain better insight into the underlying molecular mechanism of sensitivity to PRMT5 inhibition. We used T1-44 compound to inhibit PRMT5 biochemically. We found that MYCN amplified NB cancer cells were significantly more sensitive to the PRMT5 inhibition (p < 0.0062) and T1-44 treatment induced higher number of differentially expressed and alternatively spliced events in the high E2F1/MYCN expressing cell line when compared with the low E2F1/MYCN expressing cell line. We characterized differentially spliced apoptotic genes upon 200 nM treatment with T1-44 for 72 hours as these genes might have a functional impact on cell fate, we found that T1-44 induced differential splicing of apoptosis genes BCL2L11 and CDK10 in MYCN amplified compared with non-MYCN amplified NB cells. Our results highlight the role of the PRMT5, E2F1, and MYCN axis in the alternative splicing pathway and its clinical relevance in MYCN amplified NB patients. MYCN amplification is observed in many other cancer types beyond NB. Therefore, we believe that our findings will also contribute to the targeted therapy efforts in other cancer types as well.
- Published
- 2023
25. Role of PRMT1 and PRMT5 in Breast Cancer.
- Author
-
Martinez, Sébastien, Sentis, Stéphanie, Poulard, Coralie, Trédan, Olivier, and Le Romancer, Muriel
- Subjects
- *
PROTEIN arginine methyltransferases , *METASTATIC breast cancer , *BREAST cancer , *GENETIC transcription regulation , *CELL communication - Abstract
Breast cancer is the most common cancer diagnosed in women worldwide. Early-stage breast cancer is curable in ~70–80% of patients, while advanced metastatic breast cancer is considered incurable with current therapies. Breast cancer is a highly heterogeneous disease categorized into three main subtypes based on key markers orientating specific treatment strategies for each subtype. The complexity of breast carcinogenesis is often associated with epigenetic modification regulating different signaling pathways, involved in breast tumor initiation and progression, particularly by the methylation of arginine residues. Protein arginine methyltransferases (PRMT1-9) have emerged, through their ability to methylate histones and non-histone substrates, as essential regulators of cancers. Here, we present an updated overview of the mechanisms by which PRMT1 and PRMT5, two major members of the PRMT family, control important signaling pathways impacting breast tumorigenesis, highlighting them as putative therapeutic targets. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Targeting PRMT5 enhances the radiosensitivity of tumor cells grown in vitro and in vivo.
- Author
-
Degorre, Charlotte, Lohard, Steven, Bobrek, Christina N., Rawal, Komal N., Kuhn, Skyler, and Tofilon, Philip J.
- Subjects
DNA repair ,RADIATION tolerance ,DOUBLE-strand DNA breaks ,PROTEIN arginine methyltransferases ,GENETIC engineering ,CELL lines - Abstract
PRMT5 is a widely expressed arginine methyltransferase that regulates processes involved in tumor cell proliferation and survival. In the study described here, we investigated whether PRMT5 provides a target for tumor radiosensitization. Knockdown of PRMT5 using siRNA enhanced the radiosensitivity of a panel of cell lines corresponding to tumor types typically treated with radiotherapy. To extend these studies to an experimental therapeutic setting, the PRMT5 inhibitor LLY-283 was used. Exposure of the tumor cell lines to LLY-283 decreased PRMT5 activity and enhanced their radiosensitivity. This increase in radiosensitivity was accompanied by an inhibition of DNA double-strand break repair as determined by γH2AX foci and neutral comet analyses. For a normal fibroblast cell line, although LLY-283 reduced PRMT5 activity, it had no effect on their radiosensitivity. Transcriptome analysis of U251 cells showed that LLY-283 treatment reduced the expression of genes and altered the mRNA splicing pattern of genes involved in the DNA damage response. Subcutaneous xenografts were then used to evaluate the in vivo response to LLY-283 and radiation. Treatment of mice with LLY-283 decreased tumor PRMT5 activity and significantly enhanced the radiation-induced growth delay. These results suggest that PRMT5 is a tumor selective target for radiosensitization. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Sustained cancer‐relevant alternative RNA splicing events driven by PRMT5 in high‐risk neuroblastoma.
- Author
-
Bate‐Eya, Laurel Tabe, Albayrak, Gulsah, Carr, Simon Mark, Shrestha, Amit, Kanapin, Alexander, Samsonova, Anastasia, and La Thangue, Nicholas Barrie
- Subjects
- *
ALTERNATIVE RNA splicing , *RNA splicing , *GENE expression , *NEUROBLASTOMA , *PROTEIN arginine methyltransferases , *GENETIC engineering - Abstract
Protein arginine methyltransferase 5 (PRMT5) is over‐expressed in a wide variety of cancers and is implicated as having a key oncogenic role, achieved in part through its control of the master transcription regulator E2F1. We investigated the relevance of PRMT5 and E2F1 in neuroblastoma (NB) and found that elevated expression of PRMT5 and E2F1 occurs in poor prognosis high‐risk disease and correlates with an amplified Myelocytomatosis viral‐related oncogene, neuroblastoma‐derived (MYCN) gene. Our results show that MYCN drives the expression of splicing factor genes that, together with PRMT5 and E2F1, lead to a deregulated alternative RNA splicing programme that impedes apoptosis. Pharmacological inhibition of PRMT5 or inactivation of E2F1 restores normal splicing and renders NB cells sensitive to apoptosis. Our findings suggest that a sustained cancer‐relevant alternative RNA splicing programme desensitises NB cells to apoptosis, and identify PRMT5 as a potential therapeutic target for high‐risk disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Excessive fatty acids activate PRMT5/MDM2/Drosha pathway to regulate miRNA biogenesis and lipid metabolism.
- Author
-
Aijun Hou, Xiaoding Xu, Yu Zhang, Hongxiu He, Yihan Feng, Wenhui Fan, Rongrong Tan, Likun Gong, and Jing Chen
- Subjects
- *
LIPID metabolism , *FATTY acids , *NON-alcoholic fatty liver disease , *MICRORNA , *UBIQUITIN ligases - Abstract
Background: Excessive fatty acids in the liver lead to the accumulation of lipotoxic lipids and then cellular stress to further evoke the related disease, like non-alcoholic fatty liver disease (NAFLD). As reported, fatty acid stimulation can cause some specific miRNA dysregulation, which caused us to investigate the relationship between miRNA biogenesis and fatty acid overload. Methods: Gene expression omnibus (GEO) dataset analysis, miRNA-seq, miRNA cleavage assay, RT-qPCR, western blotting, immunofluorescence and co-immunoprecipitation (co-IP) were used to reveal the change of miRNAs under pathological status and explore the relevant mechanism. High fat, high fructose, high cholesterol (HFHFrHC) diet-fed mice transfected with AAV2/8-shDrosha or AAV2/8-shPRMT5 were established to investigate the in vivo effects of Drosha or PRMT5 on NAFLD phenotype. Results: We discovered that the cleavage of miRNAs was inhibited by analysing miRNA contents and detecting some representative pri-miRNAs in multiple mouse and cell models, which was further verified by the reduction of the Microprocessor activity in the presence of palmitic acid (PA). In vitro, PA could induce Drosha, the core RNase III in the Microprocessor complex, degrading through the proteasome-mediated pathway, while in vivo, knockdown of Drosha significantly promoted NAFLD to develop to a more serious stage. Mechanistically, our results demonstrated that PA can increase the methyltransferase activity of PRMT5 to degrade Drosha through MDM2, a ubiq-uitin E3 ligase for Drosha. The above results indicated that PRMT5 may be a critical regulator in lipid metabolism during NAFLD, which was confirmed by the knocking down of PRMT5 improved aberrant lipid metabolism in vitro and in vivo. Conclusions: We first demonstrated the relationship between miRNA dosage and NAFLD and proved that PA can activate the PRMT5-MDM2-Drosha signalling pathway to regulate miRNA biogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. A comprehensive investigation of PRMT5 in the prognosis and ion channel features of lung cancer
- Author
-
Yan Wang, Daifang Chu, Haichao Li, Jiangjiang Fan, Ximing Zhu, Yulong Ma, Zhongping Gu, Nianlin Xie, and Pengyu Jing
- Subjects
lung cancer ,PRMT5 ,prognosis ,epigenetics ,ion channel genes ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The increasing incidence and mortality associated with lung cancer (LC) is a significant global health challenge. The underlying mechanisms contributing to LC remain inadequately understood. However, emerging evidence suggests that the epigenetic modifier protein arginine methyltransferase 5 (PRMT5) plays a complex role in various cellular processes, including DNA repair, gene transcription, and alternative splicing, through its function in catalyzing the symmetric dimethylation of both histone and non-histone proteins. In this study, we examined the functional role of PRMT5 utilizing LC-related datasets (GSE30219, GSE50081, and TCGA LC cohort) through a series of analyses. Our findings revealed that PRMT5 was significantly overexpressed in LC samples compared to normal tissues and was correlated with overall survival and disease-free survival rates. Additionally, PRDM1 was identified as a key protein exhibiting a strong interaction with PRMT5. The prognostic model that integrated PRMT5 with clinical factors demonstrated robust performance in assessing survival outcomes. Elevated levels of PRMT5 were associated with poor prognosis in LC, as evidenced by analyses of the GSE30219, GSE50081, and TCGA-LC datasets. Furthermore, we identified 27 ion channel (IC) genes exhibited a correlation with PRMT5 in lung adenocarcinoma (LUAD), of which 9 genes were identified as statistically significant with KM survival analysis. Strikingly, all of the 9 genes, including LRRC8A, the same as PRMT5, were associated with poor prognosis in LUAD. Our research highlights the potential of PRMT5 as a novel prognostic biomarker and its relationship with IC genes in LC.
- Published
- 2024
- Full Text
- View/download PDF
30. Application and research progress of synthetic lethality in the development of anticancer therapeutic drugs
- Author
-
Xiaoliang Gong, Chunxi Liu, Haoyang Tang, Song Wu, and Qingyun Yang
- Subjects
synthetic lethality ,precision medicine ,PARP ,PRMT5 ,WRN ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
With the tremendous success of the PARP inhibitor olaparib in clinical practice, synthetic lethality has become an important field for the discovery and development of anticancer drugs. More and more synthetic lethality targets have been discovered with the rapid development of biotechnology in recent years. Currently, many drug candidates that were designed and developed on the basis of the concept of synthetic lethality have entered clinical trials. Taking representative synthetic lethal targets Poly ADP-ribose polymerase 1 (PARP1), Werner syndrome helicase (WRN) and protein arginine methyltransferase 5 (PRMT5) as examples, this article briefly discusses the application and research progress of synthetic lethality in the development of anticancer drugs.
- Published
- 2024
- Full Text
- View/download PDF
31. SCR‐7952, a highly selective MAT2A inhibitor, demonstrates synergistic antitumor activities in combination with the S‐adenosylmethionine‐competitive or the methylthioadenosine‐cooperative protein arginine methyltransferase 5 inhibitors in methylthioadenosine phosphorylase‐deleted tumors
- Author
-
Zhiyong Yu, Yi Kuang, Liting Xue, Xuan Ma, Tingting Li, Linlin Yuan, Mengying Li, Grace Xue, Zhen Li, Feng Tang, Jianxing Tang, Jinwen Shan, Weijie Wang, Renhong Tang, and Feng Zhou
- Subjects
drug combination ,MAT2A inhibitor ,MTAP‐deleted cancer ,PRMT5 ,Medicine - Abstract
Abstract The metabolic enzyme methionine adenosyltransferase 2A (MAT2A) was found to elicit synthetic lethality in methylthioadenosine phosphorylase (MTAP)‐deleted cancers, which occur in about 15% of all cancers. Here, we described a novel MAT2A inhibitor, SCR‐7952 with potent and selective antitumor effects on MTAP‐deleted cancers in both in vitro and in vivo. The cryo‐EM data indicated the high binding affinity and the allosteric binding site of SCR‐7952 on MAT2A. Different from AG‐270, SCR‐7952 exhibited little influence on metabolic enzymes and did not increase the plasma levels of bilirubin. A systematic evaluation of combination between SCR‐7952 and different types of protein arginine methyltransferase 5 (PRMT5) inhibitors indicated remarkable synergistic interactions between SCR‐7952 and the S‐adenosylmethionine‐competitive or the methylthioadenosine‐cooperative PRMT5 inhibitors, but not substrate‐competitive ones. The mechanism was via the aggravated inhibition of PRMT5 and FANCA splicing perturbations. These results indicated that SCR‐7952 could be a potential therapeutic candidate for the treatment of MTAP‐deleted cancers, both monotherapy and in combination with PRMT5 inhibitors.
- Published
- 2024
- Full Text
- View/download PDF
32. Regulation of RORα Stability through PRMT5-Dependent Symmetric Dimethylation.
- Author
-
Xiong, Gaofeng, Obringer, Brynne, Jones, Austen, Horton, Elise, and Xu, Ren
- Subjects
- *
CANCER invasiveness , *EPITHELIAL-mesenchymal transition , *RESEARCH funding , *BREAST tumors , *CELL proliferation , *TUMOR suppressor genes , *INFLAMMATION , *CELL receptors , *DEMETHYLATION , *PRECIPITIN tests - Abstract
Simple Summary: Retinoid-related orphan receptor alpha (RORα), a member of the orphan nuclear factor family, is considered a potential tumor suppressor. Our previous studies have shown that a loss of RORα is associated with enhanced breast cancer malignancy, cell invasion and proliferation, epithelial–mesenchymal transition, and cancer-associated inflammation. The mechanisms of how RORα expression is regulated in mammary epithelial cells remain incompletely understood. In this study, we revealed a direct interaction between RORα and protein arginine N-methyltransferase 5 (PRMT5), which symmetrically dimethylated the DNA-binding domain of RORα and stabilized the RORα protein. The RORα protein was decreased in PRMT5-silenced mammary epithelial cells, accompanied by enhanced invasion and migration abilities. These findings uncover a novel mechanism for RORα regulation through PRMT5-induced symmetric dimethylation in breast epithelial cells. Retinoic acid receptor-related orphan receptor alpha (RORα), a candidate tumor suppressor, is prevalently downregulated or lost in malignant breast cancer cells. However, the mechanisms of how RORα expression is regulated in breast epithelial cells remain incompletely understood. Protein arginine N-methyltransferase 5 (PRMT5), a type II methyltransferase catalyzing the symmetric methylation of the amino acid arginine in target proteins, was reported to regulate protein stability. To study whether and how PRMT5 regulates RORα, we examined the direct interaction between RORα and PRMT5 by immunoprecipitation and GST pull-down assays. The results showed that PRMT5 directly bound to RORα, and PRMT5 mainly symmetrically dimethylated the DNA-binding domain (DBD) but not the ligand-binding domain (LBD) of RORα. To investigate whether RORα protein stability is regulated by PRMT5, we transfected HEK293FT cells with RORα and PRMT5-expressing or PRMT5-silencing (shPRMT5) vectors and then examined RORα protein stability by a cycloheximide chase assay. The results showed that PRMT5 increased RORα protein stability, while silencing PRMT5 accelerated RORα protein degradation. In PRMT5-silenced mammary epithelial cells, RORα protein expression was decreased, accompanied by an enhanced epithelial–mesenchymal transition morphology and cell invasion and migration abilities. In PRMT5-overexpressed mammary epithelial cells, RORα protein was accumulated, and cell invasion was suppressed. These findings revealed a novel mechanism by which PRMT5 regulates RORα protein stability. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Alpha‐synuclein promotes PRMT5‐mediated H4R3me2s histone methylation by interacting with the BAF complex.
- Author
-
Nakamura, Takaaki, Sugeno, Naoto, Hasegawa, Takafumi, Ikeda, Kensho, Yoshida, Shun, Ishiyama, Shun, Sato, Kazuki, Takeda, Atsushi, and Aoki, Masashi
- Subjects
- *
HISTONE methylation , *CELL adhesion molecules , *NUCLEAR proteins , *ALPHA-synuclein , *PARKINSON'S disease , *IMMUNOPRECIPITATION , *CHROMATIN-remodeling complexes - Abstract
α‐Synuclein (αS) is a key molecule in the pathomechanism of Parkinson's disease. Most studies on αS to date have focused on its function in the neuronal cytosol, but its action in the nucleus has also been postulated. Indeed, several lines of evidence indicate that overexpressed αS leads to epigenomic alterations. To clarify the functional role of αS in the nucleus and its pathological significance, HEK293 cells constitutively expressing αS were used to screen for nuclear proteins that interact with αS by nanoscale liquid chromatography/tandem mass spectrometry. Interactome analysis of the 229 identified nuclear proteins revealed that αS interacts with the BRG1‐associated factor (BAF) complex, a family of multi‐subunit chromatin remodelers important for neurodevelopment, and protein arginine methyltransferase 5 (PRMT5). Subsequent transcriptomic analysis also suggested a functional link between αS and the BAF complex. Based on these results, we analyzed the effect of αS overexpression on the BAF complex in neuronally differentiated SH‐SY5Y cells and found that induction of αS disturbed the BAF maturation process, leading to a global increase in symmetric demethylation of histone H4 on arginine 3 (H4R3me2s) via enhanced BAF–PRMT5 interaction. Chromatin immunoprecipitation sequencing confirmed accumulated H4R3me2s methylation near the transcription start site of the neuronal cell adhesion molecule (NRCAM) gene, which has roles during neuronal differentiation. Transcriptional analyses confirmed the negative regulation of NRCAM by αS and PRMT5, which was reconfirmed by multiple datasets in the Gene Expression Omnibus (GEO) database. Taken together, these findings suggest that the enhanced binding of αS to the BAF complex and PRMT5 may cooperatively affect the neuronal differentiation process. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. PROGNOSTIC POTENTIAL OF PRMT5 AND DSG2 PROTEINS IN PRE-MALIGNANT CERVICAL LESIONS.
- Author
-
KRAJŇÁKOVÁ, BIBIANA, VÝBOHOVÁ, DESANKA, HURTA-CSIZMÁR, SANDRA, MEŠŤANOVÁ, VERONIKA, and ADAMKOV, MARIAN
- Abstract
Precancerous cervical lesions are metaplastic alterations of epithelial cells of the cervix, eventually developing into cervical cancer. Despite primary and secondary prevention, the burden of cervical cancer remains high globally. Protein arginine methyltransferases (PRMT) represent post-translational modifications that interact with multiple signalling pathways, playing a role in epithelial-mesenchymal transition. In complex with desmoglein-2 (DSG2), a cell adhesion protein, both participate in the progression of dysplastic changes with potential malignant development. The presented study was performed on archival paraffin-embedded blocks from adult women. The studied samples were categorised into low-grade and high-grade intraepithelial lesions. Immunohistochemical analysis was used to observe subcellular localisation, immunoreaction intensity, and percentage of PRMT5- and DSG2-expressing cells, followed by statistical analysis. Preliminary results identified statistically significant differences between the expression and subcellular localisation of proteins in question in low-grade and high-grade squamous intraepithelial lesions. The primary goal of the presented study is to perceive the involvement of PRMT5 and DSG2 in the initiation and progression of cervical lesions. Our observations indicate the potential of the assessed proteins as prognostic markers. However, further studies of PRMT5 and DSG2 are required to provide greater insight into cervical carcinogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Inhibition of PRMT5/MEP50 Arginine Methyltransferase Activity Causes Cancer Vulnerability in NDRG2 low Adult T-Cell Leukemia/Lymphoma.
- Author
-
Ichikawa, Tomonaga, Suekane, Akira, Nakahata, Shingo, Iha, Hidekatsu, Shimoda, Kazuya, Murakami, Takashi, and Morishita, Kazuhiro
- Subjects
- *
ADULT T-cell leukemia , *PROTEIN arginine methyltransferases , *ETIOLOGY of cancer , *ONCOGENIC proteins , *LYMPHOMAS , *PROTEIN stability - Abstract
N-myc downstream-regulated gene 2 (NDRG2), which is a tumour suppressor, is frequently lost in many types of tumours, including adult T-cell leukaemia/lymphoma (ATL). The downregulation of NDRG2 expression is involved in tumour progression through the aberrant phosphorylation of several important signalling molecules. We observed that the downregulation of NDRG2 induced the translocation of protein arginine methyltransferase 5 (PRMT5) from the nucleus to the cytoplasm via the increased phosphorylation of PRMT5 at Serine 335. In NDRG2low ATL, cytoplasmic PRMT5 enhanced HSP90A chaperone activity via arginine methylation, leading to tumour progression and the maintenance of oncogenic client proteins. Therefore, we examined whether the inhibition of PRMT5 activity is a drug target in NDRG2low tumours. The knockdown of PRMT5 and binding partner methylsome protein 50 (MEP50) expression significantly demonstrated the suppression of cell proliferation via the degradation of AKT and NEMO in NDRG2low ATL cells, whereas NDRG2-expressing cells did not impair the stability of client proteins. We suggest that the relationship between PRMT5/MEP50 and the downregulation of NDRG2 may exhibit a novel vulnerability and a therapeutic target. Treatment with the PRMT5-specific inhibitors CMP5 and HLCL61 was more sensitive in NDRG2low cancer cells than in NDRG2-expressing cells via the inhibition of HSP90 arginine methylation, along with the degradation of client proteins. Thus, interference with PRMT5 activity has become a feasible and effective strategy for promoting cancer vulnerability in NDRG2low ATL. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. PRMT5 activates KLF5 by methylation to facilitate lung cancer.
- Author
-
Zhou, Hai, Chang, Jing, Zhang, Jingjian, Zheng, Hongzhen, Miao, Xiang, Mo, Huimin, Sun, Jie, Jia, Qin, and Qi, Guangsheng
- Subjects
LUNG cancer ,KRUPPEL-like factors ,PROTEIN arginine methyltransferases ,CANCER cell proliferation ,METHYLATION - Abstract
The highly expressed oncogenic factor Krüppel‐like factor 5 (KLF5) promotes various cancerous processes, such as cell growth, survival, anti‐apoptosis, migration and metastasis, particularly in lung cancer. Nevertheless, the modifications to KLF5 after translation are poorly understood. Protein arginine methyltransferase 5 (PRMT5) is considered as an oncogene known to be involved in different types of carcinomas, including lung cancer. Here, we show that the expression levels of PRMT5 and KLF5 are highly expressed lung cancer. Moreover, PRMT5 interacts with KLF5 and facilitates the dimethylation of KLF5 at Arginine 41 in a manner that depends on methyltransferase activity. Downregulation or pharmaceutical suppression of PRMT5 reduces the expression of KLF5 and its downstream targets both in vitro and in vivo. Mechanistically, the dimethylation of KLF5 by PRMT5 promotes the maintenance and proliferation of lung cancer cells at least partially by stabilising KLF5 via regulation of the Akt/GSK3β signalling axis. In summary, PRMT5 methylates KLF5 to prevent its degradation, thereby promoting the maintenance and proliferation of lung cancer cells. These results suggest that targeting PRMT5/KLF5 axis may offer a potential therapeutic strategy for lung cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Exosomal circRNA RHOT1 promotes breast cancer progression by targeting miR-204-5p/ PRMT5 axis
- Author
-
Weihua Jiang, YinPing Yu, Jianghua Ou, Yongtao Li, and Ning Zhu
- Subjects
Breast Cancer ,Exosome ,CircRHOT1 ,MiR-204-5p ,PRMT5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background Circular RNA RHOT1 (circRHOT1) plays crucial roles in tumorigenesis by competing with microRNAs. It is largely abundant in tumor cell-derived exosomes. Meanwhile, cancer-derived exosomes participate in diverse biological processes. However, the expression patterns and functions of exosomal circRHOT1 in breast cancer remain unknown. This study is aimed to investigate and elucidate the exosomal circRHOT1/miR-204-5p/PRMT5 axis in breast cancer. Methods The exosomes derived from serum samples of breast cancer patients and breast cancer cell lines were characterized using transmission electron microscopy and Western blot. MTT, colony formation, wound healing, and transwell assays were utilized to analyze cell proliferation, migration, and invasion of breast cancer cells. Flow cytometry was used for apoptosis analysis. The bioinformatics method was employed to screen differentially expressed novel circRNAs and predict the microRNA targets of circRHOT1. Dual-luciferase reporter gene assays were performed to verify their direct interaction. Finally, Xenograft experiments were used to investigate the effect of exosomal circRHOT1 on tumor growth in vivo. Results CircRHOT1 exhibited significantly high expression in exosomes derived from the serum of breast cancer patients and breast cancer cell lines, which suggested its potential diagnostic value. Breast cancer-derived exosomes promoted the cell proliferation, migration, invasion, and epithelial-mesenchymal transition of breast cancer cells while inhibiting apoptosis. However, exosomes with downregulated circRHOT1 inhibited the growth of co-cultured cells. Mechanistically, circRHOT1 acted as a sponge of miR-204-5p and promoted protein arginine methyltransferase 5 (PRMT5) expression. Moreover, miR-204-5p inhibitor and pcPRMT5 could reverse the tumor suppressive effects mediated by circRHOT1-knockdown. Furthermore, treatment with exosomes derived from breast cancer cells with circRHOT1 knockdown attenuated tumor growth in tumor-bearing nude mice, which was accompanied by a reduction in PRMT5 expression and an enhancement of miR-204-5p expression. Conclusion The exosomal circRHOT1 may promote breast cancer progression by regulating the miR-204-5p/PRMT5 axis. The current study strengthens the role of circRHOT1, miR-204-5p, and PRMT5 in breast cancer development and provides a potential treatment strategy for breast cancer.
- Published
- 2023
- Full Text
- View/download PDF
38. Carcinogenesis and Prognostic Utility of Arginine Methylation-Related Genes in Hepatocellular Cancer
- Author
-
Waleed Ali, Weirui Xiao, Henry Hoang, Vincent Cali, and Andre Kajdacsy-Balla
- Subjects
arginine methylation ,hepatocellular carcinoma ,TCGA ,PRMT5 ,in silico ,Biology (General) ,QH301-705.5 - Abstract
Protein arginine methylation is among the most important post-translational modifications and has been studied in cancers such as those of the lung and breast. However, comparatively less has been investigated regarding hepatocellular carcinoma, with an annual incidence of almost one million cases. Through using in silico methods, this study examined arginine methylation-related gene expression and methylation levels, and alongside network and enrichment analysis attempted to find how said genes can drive tumorigenesis and offer possible therapeutic targets. We found a robust relationship among the selected methylation genes, with ⅞ showing prognostic value regarding overall survival, and a medley of non-arginine methylation pathways also being highlighted through the aforementioned analysis. This study furthers our knowledge of the methylation and expression patterns of arginine histone methylation-related genes, offering jumping points for further wet-lab studies.
- Published
- 2023
- Full Text
- View/download PDF
39. Genetic Alterations of NF-κB and Its Regulators: A Rich Platform to Advance Colorectal Cancer Diagnosis and Treatment.
- Author
-
Alipourgivi, Faranak, Motolani, Aishat, Qiu, Alice Y., Qiang, Wenan, Yang, Guang-Yu, Chen, Shuibing, and Lu, Tao
- Subjects
- *
HUMAN embryonic stem cells , *COLORECTAL cancer , *CANCER treatment , *PLURIPOTENT stem cells , *CANCER diagnosis - Abstract
Colorectal cancer (CRC) is the third leading cause of cancer mortality in the United States, with an estimated 52,000 deaths in 2023. Though significant progress has been made in both diagnosis and treatment of CRC in recent years, genetic heterogeneity of CRC—the culprit for possible CRC relapse and drug resistance, is still an insurmountable challenge. Thus, developing more effective therapeutics to overcome this challenge in new CRC treatment strategies is imperative. Genetic and epigenetic changes are well recognized to be responsible for the stepwise development of CRC malignancy. In this review, we focus on detailed genetic alteration information about the nuclear factor (NF)-κB signaling, including both NF-κB family members, and their regulators, such as protein arginine methyltransferase 5 (PRMT5), and outer dynein arm docking complex subunit 2 (ODAD2, also named armadillo repeat-containing 4, ARMC4), etc., in CRC patients. Moreover, we provide deep insight into different CRC research models, with a particular focus on patient-derived xenografts (PDX) and organoid models, and their potential applications in CRC research. Genetic alterations on NF-κB signaling components are estimated to be more than 50% of the overall genetic changes identified in CRC patients collected by cBioportal for Cancer Genomics; thus, emphasizing its paramount importance in CRC progression. Consequently, various genetic alterations on NF-κB signaling may hold great promise for novel therapeutic development in CRC. Future endeavors may focus on utilizing CRC models (e.g., PDX or organoids, or isogenic human embryonic stem cell (hESC)-derived colonic cells, or human pluripotent stem cells (hPSC)-derived colonic organoids, etc.) to further uncover the underpinning mechanism of these genetic alterations in NF-κB signaling in CRC progression. Moreover, establishing platforms for drug discovery in dishes, and developing Biobanks, etc., may further pave the way for the development of innovative personalized medicine to treat CRC in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. PRMT5 participates in B cell overactivation in patients with primary Sjogren's syndrome (pSS) through RSAD2‐mediated NF‐κB signaling.
- Author
-
Zhu, Hong, Zheng, Jian, Zhou, Yan, Wu, Tong, and Zhu, Tiantian
- Subjects
- *
SJOGREN'S syndrome , *B cells , *IMMUNOGLOBULIN M , *PROTEIN arginine methyltransferases , *CD19 antigen , *IMMUNOGLOBULINS - Abstract
Objective: There are new evidences that protein arginine methyltransferase 5 (PRMT5) is widely involved in the progression of various diseases, but its effect is unclear on Primary Sjogren's syndrome (pSS). The main purpose of this study is to explore the regulatory effect of PRMT5 on pSS and its potential mechanisms. Methods: CD40L treated CD19 + B cells to construct a cell model of pSS. CCK‐8 assay and Annexin V‐FITC/PI kits were used to measure cell proliferation and apoptosis. ELISA assay was used to determine the contents of IL‐6 and TNF‐α in CD19 + B cells. And commercial kits were used to detect the levels of immunoglobins (IgG, IgM, and IgA) in CD40L‐treated CD19 + B cells. And successfully constructed a pSS mouse model. Results: The results revealed an increase in the expression of PRMT5 in CD19 + B cells from patients with pSS. After CD40L treatment, the knockdown of PRMT5 prominently decreased cell viability, the production level of immunoglobulins (IgG, IgM, and IgA), and the content of IL‐10, increased the content of IL‐6 and IL‐8, and promoted the apoptosis of pSS CD19 + B cells. Mechanistically, PRMT5 negatively regulated the RSAD2 and nuclear factor kappa‐B (NF‐κB) signaling pathway. Furthermore, overexpression of RSAD2 and p65 significantly rescued the effect of PRMT5 knockdown on proliferation, immunoglobin production and secreting cytokines in CD40L‐treated CD19 + B cells. More importantly, inhibition of PRMT5 significantly inhibited the symptoms of pSS mice. Conclusions: Low‐expression of PRMT5 through inactivation of RSAD2/NF‐κB signalling pathway alleviates the hyperactivity of B cells, which may provide theoretical basis and potential therapeutic targets for clinical treatment of pSS. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. MST2 methylation by PRMT5 inhibits Hippo signaling and promotes pancreatic cancer progression.
- Author
-
Sun, Yan, Jin, Xin, Meng, Junpeng, Guo, Feng, Chen, Taoyu, Zhao, Xiaoyan, Wu, Heshui, and Ren, Dianyun
- Subjects
- *
HIPPO signaling pathway , *PANCREATIC cancer , *CANCER invasiveness , *PROTEIN arginine methyltransferases , *YAP signaling proteins , *VIRUS inactivation - Abstract
The Hippo signaling axis is a tumor suppressor pathway that is activated by various extra‐pathway factors to regulate cell differentiation and organ development. Recent studies have reported that autophosphorylation of the core kinase cassette stimulates activation of the Hippo signaling cascade. Here, we demonstrate that protein arginine methyltransferase 5 (PRMT5) contributes to inactivation of the Hippo signaling pathway in pancreatic cancer. We show that the Hippo pathway initiator serine/threonine kinase 3 (STK3, also known as MST2) of Hippo signaling pathway can be symmetrically di‐methylated by PRMT5 at arginine‐461 (R461) and arginine‐467 (R467) in its SARAH domain. Methylation suppresses MST2 autophosphorylation and kinase activity by blocking its homodimerization, thereby inactivating Hippo signaling pathway in pancreatic cancer. Moreover, we also show that the specific PRMT5 inhibitor GSK3326595 re‐activates the dysregulated Hippo signaling pathway and inhibits the growth of human pancreatic cancer xenografts in immunodeficient mice, thus suggesting potential clinical application of PRMT5 inhibitors in pancreatic cancer. Synopsis: Activation of the YAP transcriptional co‐activator, frequently occurring in pancreatic cancer, is suppressed by the Hippo pathway initiated by the MST2 kinase. Here, arginine methylation via PRMT5 is found to inhibit MST2 activation and may present a target for cancer treatment. MST2 is dimethylated at R461 and R467 in its C‐terminal SARAH domain.MST2 dimethylation decreases its homodimerization, suppressing MST2 autophosphorylation and kinase activity.PRMT5 catalyzes the symmetrical SARAH domain dimethylation of MST2.The PRMT5 inhibitor GSK3326595 re‐activates Hippo signaling in human pancreatic cancer‐derived xenografts. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
42. Prmt5 deficiency inhibits CD4+ T-cell Klf2/S1pr1 expression and ameliorates EAE disease
- Author
-
Yingxia Zheng, Zheyi Chen, Bingqian Zhou, Shiyu Chen, Ningdai Chen, and Lisong Shen
- Subjects
Prmt5 ,S1pr1 ,Klf2 ,Migration ,EAE ,CD4+ T ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Protein arginine methyltransferase 5 (Prmt5) is the main type II methyltransferase, catalyzes protein arginine residue symmetric dimethylation, and modulates normal cellular physiology and disease progression. Prmt5 inhibition or deletion in CD4+ T cells has been reported to ameliorate experimental autoimmune encephalomyelitis (EAE), but the detailed molecular mechanisms have not yet been elucidated. Methods EAE was induced by administration of myelin oligodendrocyte glycoprotein (MOG35–55) in T cells Prmt5 conditional knockout (CD4-cre-Prmt5fl/fl, Prmt5cko) and Prmt5fl/fl (WT) mice. Flow cytometry, single-cell RNA sequencing, ATAC sequencing and chromatin immunoprecipitation assay (ChIP) approaches were used to explore the detail mechanisms. Results We find that Prmt5cko mice are resistant to EAE; infiltrating inflammatory CD4+ T cells in the central nervous system (CNS) are greatly reduced. However, in Prmt5cko mice, T cells in the spleen show much more proliferation and activation properties, the total number of CD4+ T cells in the spleen is not reduced, and the percentage of Rora+ CD4+ T cells is elevated. Also, CD4+ T cells express lower levels of S1pr1 and Klf2 than WT mice, which may influence pathogenic CD4+ T-cell egress from the spleen and migration to the CNS. Moreover, the single-cell ATAC sequence and ChIP assay reveal that the transcription factor Klf2 is enriched at the S1pr1 promoter and that Klf2 motif activity is reduced in Prmt5cko mice. Conclusions Our study delineates the undiscovered role of Prmt5 in T-cell biology in which Prmt5 may inhibit Klf2-S1pr1 pathway to ameliorate EAE disease. Controlling T-cell Prmt5 expression may be helpful for the treatment of autoimmune diseases.
- Published
- 2023
- Full Text
- View/download PDF
43. Nuclear PRMT5 is a biomarker of sensitivity to tamoxifen in ERα+ breast cancer
- Author
-
Coralie Poulard, Thuy Ha Pham, Youenn Drouet, Julien Jacquemetton, Ausra Surmielova, Loay Kassem, Benoite Mery, Christine Lasset, Jonathan Reboulet, Isabelle Treilleux, Elisabetta Marangoni, Olivier Trédan, and Muriel Le Romancer
- Subjects
arginine methylation ,estrogen receptor ,PRMT5 ,resistance ,tamoxifen ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Endocrine therapies targeting estrogen signaling, such as tamoxifen, have significantly improved management of estrogen receptor alpha (ERα)‐positive breast cancers. However, their efficacy is limited by intrinsic and acquired resistance to treatment, and there is currently no predictive marker of response to these anti‐estrogens to guide treatment decision. Here, using two independent cohorts of breast cancer patients, we identified nuclear PRMT5 expression as an independent predictive marker of sensitivity to tamoxifen. Mechanistically, we discovered that tamoxifen stimulates ERα methylation by PRMT5, a key event for its binding to corepressors such as SMRT and HDAC1, participating in the inhibition of the transcriptional activity of ERα. Although PRMT5 is mainly localized in the cytoplasm of tumor cells, our analyses show that tamoxifen triggers its nuclear translocation in tamoxifen‐sensitive tumors but not in resistant ones. Hence, we unveil a biomarker of sensitivity to tamoxifen in ERα‐positive breast tumors that could be used to enhance the response of breast cancer patients to endocrine therapy, by fostering its nuclear expression.
- Published
- 2023
- Full Text
- View/download PDF
44. PRMT5 promotes chemotherapy‐induced neuroendocrine differentiation in NSCLC
- Author
-
Qi Shen, Yi Liu, Xuehong Deng, and Chang‐Deng Hu
- Subjects
acquired resistance ,chemotherapy ,NED ,neuroendocrine differentiation ,PRMT5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background In response to therapeutic treatments, cancer cells can exhibit a variety of resistance phenotypes including neuroendocrine differentiation (NED). NED is a process by which cancer cells can transdifferentiate into neuroendocrine‐like cells in response to treatments, and is now widely accepted as a key mechanism of acquired therapy resistance. Recent clinical evidence has suggested that non‐small cell lung cancer (NSCLC) can also transform into small cell lung cancer (SCLC) in patients treated with EGFR inhibitors. However, whether chemotherapy induces NED to confer therapy resistance in NSCLC remains unknown. Methods We evaluated whether NSCLC cells can undergo NED in response to chemotherapeutic agents etoposide and cisplatin. By Knock‐down of PRMT5 or pharmacological inhibition of PRMT5 to identify its role in the NED process. Results We observed that both etoposide and cisplatin can induce NED in multiple NSCLC cell lines. Mechanistically, we identified protein arginine methyltransferase 5 (PRMT5) as a critical mediator of chemotherapy‐induced NED. Significantly, the knock‐down of PRMT5 or pharmacological inhibition of PRMT5 suppressed the induction of NED and increased the sensitivity to chemotherapy. Conclusion Taken together, our results suggest that targeting PRMT5 may be explored as a chemosensitization approach by inhibiting chemotherapy‐induced NED.
- Published
- 2023
- Full Text
- View/download PDF
45. The crescent-like Golgi ribbon is shaped by the Ajuba/PRMT5/Aurora-A complex-modified HURP
- Author
-
Shao-Chih Chiu, Xin-Ting Yang, Tong-You Wade Wei, Yu-Ting Amber Liao, Jo-Mei Maureen Chen, Yi-Chun Kuo, Chun-Chih Jared Liu, Chiao-Yun Cheng, Yu-Ting Jenny Huang, Yun-Ru Jaoying Huang, He-Lian Joe Wu, Chang-Xin Wan, Jia-Rung Tsai, and Chang-Tze Ricky Yu
- Subjects
Golgi ribbon ,HURP ,Ajuba ,PRMT5 ,Aurora-A ,ARF1 ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Golgi apparatus (GA) is assembled as a crescent-like ribbon in mammalian cells under immunofluorescence microscope without knowing the shaping mechanisms. It is estimated that roughly 1/5 of the genes encoding kinases or phosphatases in human genome participate in the assembly of Golgi ribbon, reflecting protein modifications play major roles in building Golgi ribbon. Methods To explore how Golgi ribbon is shaped as a crescent-like structure under the guidance of protein modifications, we identified a protein complex containing the scaffold proteins Ajuba, two known GA regulators including the protein kinase Aurora-A and the protein arginine methyltransferase PRMT5, and the common substrate of Aurora-A and PRMT5, HURP. Mutual modifications and activation of PRMT5 and Aurora-A in the complex leads to methylation and in turn phosphorylation of HURP, thereby producing HURP p725. The HURP p725 localizes to GA vicinity and its distribution pattern looks like GA morphology. Correlation study of the HURP p725 statuses and GA structure, site-directed mutagenesis and knockdown-rescue experiments were employed to identify the modified HURP as a key regulator assembling GA as a crescent ribbon. Results The cells containing no or extended distribution of HURP p725 have dispersed GA membranes or longer GA. Knockdown of HURP fragmentized GA and HURP wild type could, while its phosphorylation deficiency mutant 725A could not, restore crescent Golgi ribbon in HURP depleted cells, collectively indicating a crescent GA-constructing activity of HURP p725. HURP p725 is transported, by GA membrane-associated ARF1, Dynein and its cargo adaptor Golgin-160, to cell center where HURP p725 forms crescent fibers, binds and stabilizes Golgi assembly factors (GAFs) including TRIP11, GRASP65 and GM130, thereby dictating the formation of crescent Golgi ribbon at nuclear periphery. Conclusions The Ajuba/PRMT5/Aurora-A complex integrates the signals of protein methylation and phosphorylation to HURP, and the HURP p725 organizes GA by stabilizing and recruiting GAFs to its crescent-like structure, therefore shaping GA as a crescent ribbon. Therefore, the HURP p725 fiber serves a template to construct GA according to its shape. Video Abstract
- Published
- 2023
- Full Text
- View/download PDF
46. PRMT5 regulates the polysaccharide content by controlling the splicing of thaumatin-like protein in Ganoderma lucidum
- Author
-
Rui Liu, Zhengyan Yang, Tao Yang, Zi Wang, Xin Chen, Jing Zhu, Ang Ren, Liang Shi, Hanshou Yu, and Mingwen Zhao
- Subjects
PRMT5 ,alternative splicing ,thaumatin-like protein ,G. lucidum polysaccharide ,Microbiology ,QR1-502 - Abstract
ABSTRACT In the current study, the silencing of protein arginine methyltransferase 5 (PRMT5) decreased the polysaccharide content of Ganoderma lucidum compared to that of the wild-type (WT) strain. Furthermore, RNA-seq screening showed that the glycan degradation process-related gene thaumatin-like protein (TLP) was alternatively spliced in WT strains via retained introns, leading to the production of a longer TLP1 isoform and a shorter TLP2 isoform; however, only the TLP2 isoform was observed in PRMT5i strains. Experiments examining the polysaccharide content of the TLP silencing, TLP1 overexpression (OE-TLP1), and TLP2 overexpression (OE-TLP2) transformants revealed that TLP2 plays a more important role than TLP1 in polysaccharide degradation. Through a combination of yeast two-hybrid, bimolecular fluorescence complementation and surface plasmon resonance assays, we found that TLP2 directly physically interacted with phosphoglucose isomerase (PGI), a key enzyme in polysaccharide synthesis, and thereby increased PGI activity. However, TLP1 failed to interact with PGI, and PGI activity was not affected. Further inspection showed that the polysaccharide content was decreased in the OE-TLP2 strains but not significantly changed in the OE-TLP1 strains compared with that in the WT strains. In addition, the polysaccharide content of the PRMT5-TLP-cosilenced strains was not significantly different from that of the WT strains. These results demonstrate that PRMT5 modulates TLP processing of pre-mRNA transcripts and thereby decreases the polysaccharide content. IMPORTANCE PRMT5 contributes to secondary metabolite biosynthesis in Ganoderma lucidum. However, the mechanism through which PRMT5 regulates the biosynthesis of secondary metabolites remains unclear. In the current study, PRMT5 silencing led to a significant decrease in the biosynthesis of polysaccharides from G. lucidum through the action of the alternative splicing of TLP. A shorter TLP2 isoform can directly bind to PGI and regulated polysaccharide biosynthesis. These results suggest that PRMT5 enhances PGI activity by regulating TLP binding to PGI. The results of the current study reveal a novel target gene for PRMT5-mediated alternative splicing and provide a reference for the identification of PRMT5 regulatory target genes.
- Published
- 2023
- Full Text
- View/download PDF
47. Pan-cancer analysis identifies protein arginine methyltransferases PRMT1 and PRMT5 and their related signatures as markers associated with prognosis, immune profile, and therapeutic response in lung adenocarcinoma
- Author
-
Jia Wang, Meng Wu, Jujie Sun, Minxin Chen, Zengfu Zhang, Jinming Yu, and Dawei Chen
- Subjects
PRMT1 ,PRMT5 ,Prognosis ,Tumor immune microenvironment ,Type I interferon pathway ,Radiotherapy ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Purpose: Protein arginine methyltransferases (PRMTs) regulate several signal transduction pathways involved in cancer progression. Recently, it has been reported that PRMTs are closely related to anti-tumor immunity; however, the underlying mechanisms have yet to be studied in lung adenocarcinoma (LUAD). In this study, we focused on PRMT1 and PRMT5, key members of the PRMT family. And their signatures in lung carcinoma associated with prognosis, immune profile, and therapeutic response including immunotherapy and radiotherapy were explored. Methods: To understand the function of PRMT1 and PRMT5 in tumor cells, we examined the association between the expression of PRMT1 and PRMT5 and the clinical, genomic, and immune characteristics, as well as the sensitivity to immunotherapy and radiotherapy. Specifically, our investigation focused on the role of PRMT1 and PRMT5 in tumor progression, with particular emphasis on interferon-stimulated genes (ISGs) and the pathway of type I interferon. Furthermore, the influence of proliferation, migration, and invasion ability was investigated based on the expression of PRMT1 and PRMT5 in human lung adenocarcinoma cell lines. Results: Through the examination of receiver operating characteristic (ROC) and survival studies, PRMT1 and PRMT5 were identified as potential biomarkers for the diagnosis and prognosis. Additionally, heightened expression of PRMT1 or PRMT5 was associated with immunosuppressive microenvironments. Furthermore, a positive correlation was observed between the presence of PRMT1 or PRMT5 with microsatellite instability, tumor mutational burden, and neoantigens in the majority of cancers. Moreover, the predictive potential of PRMT1 or PRMT5 in individuals undergoing immunotherapy has been acknowledged. Our study ultimately revealed that the inhibition of PRMT1 and PRMT5 in lung adenocarcinoma resulted in the activation of the cGAS-STING pathway, especially after radiation. Favorable prognosis was observed in lung adenocarcinoma patients receiving radiotherapy with reduced PRMT1 or PRMT5 expression. It was also found that the expression of PRMT1 and PRMT5 influenced proliferation, migration, and invasion of human lung adenocarcinoma cell lines. Conclusion: The findings indicate that PRMT1 and PRMT5 exhibit potential as immune-related biomarkers for the diagnosis and prognosis of cancer. Furthermore, these biomarkers could be therapeutically targeted to augment the efficacy of immunotherapy and radiotherapy in lung adenocarcinoma.
- Published
- 2023
- Full Text
- View/download PDF
48. Exosomal circRNA RHOT1 promotes breast cancer progression by targeting miR-204-5p/ PRMT5 axis.
- Author
-
Jiang, Weihua, Yu, YinPing, Ou, Jianghua, Li, Yongtao, and Zhu, Ning
- Subjects
BREAST cancer ,CANCER cell growth ,CANCER invasiveness ,EXOSOMES ,CIRCULAR RNA ,PROTEIN arginine methyltransferases - Abstract
Background: Circular RNA RHOT1 (circRHOT1) plays crucial roles in tumorigenesis by competing with microRNAs. It is largely abundant in tumor cell-derived exosomes. Meanwhile, cancer-derived exosomes participate in diverse biological processes. However, the expression patterns and functions of exosomal circRHOT1 in breast cancer remain unknown. This study is aimed to investigate and elucidate the exosomal circRHOT1/miR-204-5p/PRMT5 axis in breast cancer. Methods: The exosomes derived from serum samples of breast cancer patients and breast cancer cell lines were characterized using transmission electron microscopy and Western blot. MTT, colony formation, wound healing, and transwell assays were utilized to analyze cell proliferation, migration, and invasion of breast cancer cells. Flow cytometry was used for apoptosis analysis. The bioinformatics method was employed to screen differentially expressed novel circRNAs and predict the microRNA targets of circRHOT1. Dual-luciferase reporter gene assays were performed to verify their direct interaction. Finally, Xenograft experiments were used to investigate the effect of exosomal circRHOT1 on tumor growth in vivo. Results: CircRHOT1 exhibited significantly high expression in exosomes derived from the serum of breast cancer patients and breast cancer cell lines, which suggested its potential diagnostic value. Breast cancer-derived exosomes promoted the cell proliferation, migration, invasion, and epithelial-mesenchymal transition of breast cancer cells while inhibiting apoptosis. However, exosomes with downregulated circRHOT1 inhibited the growth of co-cultured cells. Mechanistically, circRHOT1 acted as a sponge of miR-204-5p and promoted protein arginine methyltransferase 5 (PRMT5) expression. Moreover, miR-204-5p inhibitor and pcPRMT5 could reverse the tumor suppressive effects mediated by circRHOT1-knockdown. Furthermore, treatment with exosomes derived from breast cancer cells with circRHOT1 knockdown attenuated tumor growth in tumor-bearing nude mice, which was accompanied by a reduction in PRMT5 expression and an enhancement of miR-204-5p expression. Conclusion: The exosomal circRHOT1 may promote breast cancer progression by regulating the miR-204-5p/PRMT5 axis. The current study strengthens the role of circRHOT1, miR-204-5p, and PRMT5 in breast cancer development and provides a potential treatment strategy for breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. The potential and challenges of targeting MTAP-negative cancers beyond synthetic lethality.
- Author
-
Bray, Chandler, Balcells, Cristina, McNeish, Iain A., and Keun, Hector C.
- Subjects
TUMOR suppressor genes ,METHYLTRANSFERASES ,PROTEIN arginine methyltransferases ,PHARMACODYNAMICS ,ADENOSYLMETHIONINE ,HISTONES - Abstract
Approximately 15% of cancers exhibit loss of the chromosomal locus 9p21.3 -- the genomic location of the tumour suppressor gene CDKN2A and the methionine salvage gene methylthioadenosine phosphorylase (MTAP). A loss of MTAP increases the pool of its substrate methylthioadenosine (MTA), which binds to and inhibits activity of protein arginine methyltransferase 5 (PRMT5). PRMT5 utilises the universal methyl donor S-adenosylmethionine (SAM) to methylate arginine residues of protein substrates and regulate their activity, notably histones to regulate transcription. Recently, targeting PRMT5, or MAT2A that impacts PRMT5 activity by producing SAM, has shown promise as a therapeutic strategy in oncology, generating synthetic lethality in MTAPnegative cancers. However, clinical development of PRMT5 and MAT2A inhibitors has been challenging and highlights the need for further understanding of the downstream mediators of drug effects. Here, we discuss the rationale and methods for targeting the MAT2A/PRMT5 axis for cancer therapy. We evaluate the current limitations in our understanding of the mechanism of MAT2A/PRMT5 inhibitors and identify the challenges that must be addressed to maximise the potential of these drugs. In addition, we review the current literature defining downstream effectors of PRMT5 activity that could determine sensitivity to MAT2A/PRMT5 inhibition and therefore present a rationale for novel combination therapies that may not rely on synthetic lethality with MTAP loss. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
50. The PRMT5 inhibitor C9 mitigates hypoxia‐induced carboplatin resistance in lung cancer by inducing autophagy.
- Author
-
Fan, Jiangjiang, Li, Haichao, Ruan, Qiong, Zhu, Ximing, Jing, Pengyu, and Gu, Zhongping
- Subjects
- *
CARBOPLATIN , *LUNG cancer , *AUTOPHAGY , *CANCER cells , *CARCINOGENESIS , *CANCER chemotherapy - Abstract
Hypoxia, a common feature of solid tumors, can promote chemoresistance in cancer cells. PRMT5 mediates various cellular processes involved in cancer development and progression. However, the role of PRMT5 in hypoxia‐induced chemoresistance is unclear. In this study, hypoxia upregulated PRMT5 expression in lung cancer cells. Additionally, PRMT5 overexpression promoted cancer cell resistance to carboplatin. In carboplatin‐resistant cancer cells, PRMT5 overexpression promoted the methylation of ULK1, a critical regulator of autophagy. ULK1 hypermethylation leads to the upregulation of autophagy, which can improve the survival of cancer cells under hypoxic conditions. Furthermore, this study demonstrated that the PRMT5 inhibitor C9 significantly enhanced the sensitivity of lung cancer cells to carboplatin. These findings suggest that targeting PRMT5‐mediated autophagy with C9 can overcome hypoxia‐induced carboplatin resistance and improve the efficacy of chemotherapy in patients with cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.