1
|
Martinez S, Sentis S, Poulard C, Trédan O, Le Romancer M. Role of PRMT1 and PRMT5 in Breast Cancer. Int J Mol Sci 2024; 25:8854. [PMID: 39201539 PMCID: PMC11354362 DOI: 10.3390/ijms25168854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
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.
Collapse
Affiliation(s)
- Sébastien Martinez
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
| | - Stéphanie Sentis
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
| | - Coralie Poulard
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
| | - Olivier Trédan
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
- Oncology Department, Centre Leon Bérard, F-69008 Lyon, France
| | - Muriel Le Romancer
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Claude Bernard Lyon 1, F-69000 Lyon, France
| |
Collapse
|
2
|
Zhang Y, Wei S, Jin EJ, Jo Y, Oh CM, Bae GU, Kang JS, Ryu D. Protein Arginine Methyltransferases: Emerging Targets in Cardiovascular and Metabolic Disease. Diabetes Metab J 2024; 48:487-502. [PMID: 39043443 PMCID: PMC11307121 DOI: 10.4093/dmj.2023.0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 06/17/2024] [Indexed: 07/25/2024] Open
Abstract
Cardiovascular diseases (CVDs) and metabolic disorders stand as formidable challenges that significantly impact the clinical outcomes and living quality for afflicted individuals. An intricate comprehension of the underlying mechanisms is paramount for the development of efficacious therapeutic strategies. Protein arginine methyltransferases (PRMTs), a class of enzymes responsible for the precise regulation of protein methylation, have ascended to pivotal roles and emerged as crucial regulators within the intrinsic pathophysiology of these diseases. Herein, we review recent advancements in research elucidating on the multifaceted involvements of PRMTs in cardiovascular system and metabolic diseases, contributing significantly to deepen our understanding of the pathogenesis and progression of these maladies. In addition, this review provides a comprehensive analysis to unveil the distinctive roles of PRMTs across diverse cell types implicated in cardiovascular and metabolic disorders, which holds great potential to reveal novel therapeutic interventions targeting PRMTs, thus presenting promising perspectives to effectively address the substantial global burden imposed by CVDs and metabolic disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, Korea
| | - Shibo Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Eun-Ju Jin
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Gyu-Un Bae
- Muscle Physiome Institute, College of Pharmacy, Sookmyung Women’s University, Seoul, Korea
- Research Institute of Aging-Related Diseases, AniMusCure Inc., Suwon, Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, Suwon, Korea
- Research Institute of Aging-Related Diseases, AniMusCure Inc., Suwon, Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Korea
| |
Collapse
|
3
|
Zhou S, Zhang Q, Yang H, Zhu Y, Hu X, Wan G, Yu L. Targeting type I PRMTs as promising targets for the treatment of pulmonary disorders: Asthma, COPD, lung cancer, PF, and PH. Life Sci 2024; 342:122538. [PMID: 38428571 DOI: 10.1016/j.lfs.2024.122538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
Pulmonary disorders, including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), pulmonary hypertension (PH), and lung cancer, seriously impair the quality of lives of patients. A deeper understanding of the occurrence and development of the above diseases may inspire new strategies to remedy the scarcity of treatments. Type I protein arginine methyltransferases (PRMTs) can affect processes of inflammation, airway remodeling, fibroblast proliferation, mitochondrial mass, and epithelial dysfunction through substrate methylation and non-enzymatic activity, thus affecting the occurrence and development of asthma, COPD, lung cancer, PF, and PH. As potential therapeutic targets, inhibitors of type I PRMTs are developed, moreover, representative compounds such as GSK3368715 and MS023 have also been used for early research. Here, we collated structures of type I PRMTs inhibitors and compared their activity. Finally, we highlighted the physiological and pathological associations of type I PRMTs with asthma, COPD, lung cancer, PF, and PH. The developing of type I PRMTs modulators will be beneficial for the treatment of these diseases.
Collapse
Affiliation(s)
- Shuyan Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiangsheng Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Honglin Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongxia Zhu
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guoquan Wan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luoting Yu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Peng L, Sun X, Yi X, Wang Z, Chen K. EGR3 reduces podocyte inflammatory damage in obesity related glomerulopathy by inhibiting the PRMT1 /p -STAT3 pathway. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:349-358. [PMID: 38970508 PMCID: PMC11208401 DOI: 10.11817/j.issn.1672-7347.2024.230394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Indexed: 07/08/2024]
Abstract
OBJECTIVES Obesity related glomerulopathy (ORG) is induced by obesity, but the pathogenesis remains unclear. This study aims to investigate the expression of early growth response protein 3 (EGR3) in the renal cortex tissues of ORG patients and high-fat diet-induced obese mice, and to further explore the molecular mechanism of EGR3 in inhibiting palmitic acid (PA) induced human podocyte inflammatory damage. METHODS Renal cortex tissues were collected from ORG patients (n=6) who have been excluded from kidney damage caused by other diseases and confirmed by histopathology, and from obese mice induced by high-fat diet (n=10). Human and mouse podocytes were intervened with 150 μmol/L PA for 48 hours. EGR3 was overexpressed or silenced in human podocytes. Enzyme linked immunosorbent assay (ELISA) was used to detcet the levels of interleukin-6 (IL-6) and interleukin-1β (IL-1β). Real-time RT-PCR was used to detect the mRNA expressions of EGR3, podocytes molecular markers nephrosis 1 (NPHS1), nephrosis 2 (NPHS2), podocalyxin (PODXL), and podoplanin (PDPN). RNA-seq was performed to detect differentially expressed genes (DEGs) after human podocytes overexpressing EGR3 and treated with 150 μmol/L PA compared with the control group. Co-immunoprecipitation (Co-IP) combined with liquid chromatography tandem mass spectrometry (LC-MS) was used to detect potential interacting proteins of EGR3 and the intersected with the RNA-seq results. Co-IP confirmed the interaction between EGR3 and protein arginine methyltransferases 1 (PRMT1), after silencing EGR3 and PRMT1 inhibitor intervention, the secretion of IL-6 and IL-1β in PA-induced podocytes was detected. Western blotting was used to detect the expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) after overexpression or silencing of EGR3. RESULTS EGR3 was significantly upregulated in renal cortex tissues of ORG patients and high-fat diet-induced obese mice (both P<0.01). In addition, after treating with 150 μmol/L PA for 48 hours, the expression of EGR3 in human and mouse podocytes was significantly upregulated (both P<0.05). Overexpression or silencing of EGR3 in human podocytes inhibited or promoted the secretion of IL-6 and IL-1β in the cell culture supernatant after PA intervention, respectively, and upregulated or downregulated the expression of NPHS1, PODXL, NPHS2,and PDPN (all P<0.05). RNA-seq showed a total of 988 DEGs, and Co-IP+LC-MS identified a total of 238 proteins that may interact with EGR3. Co-IP confirmed that PRMT1 was an interacting protein with EGR3. Furthermore, PRMT1 inhibitors could partially reduce PA-induced IL-6 and IL-1β secretion after EGR3 silencing in human podocytes (both P<0.05). Overexpression or silencing of EGR3 negatively regulated the expression of PRMT1 and p-STAT3. CONCLUSIONS EGR3 may reduce ORG podocyte inflammatory damage by inhibiting the PRMT1/p-STAT3 pathway.
Collapse
Affiliation(s)
- Lin Peng
- Department of Nephrology, First Hospital of Changsha, Changsha 410005.
| | - Xiaoying Sun
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xuan Yi
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhouqi Wang
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ke Chen
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
5
|
Chen T, Liu J, Li S, Wang P, Shang G. The role of protein arginine N-methyltransferases in inflammation. Semin Cell Dev Biol 2024; 154:208-214. [PMID: 36075843 DOI: 10.1016/j.semcdb.2022.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022]
Abstract
Protein arginine methyltransferases (PRMTs) promote the methylation of numerous proteins at their arginine residues. An increasing number of publications have suggested that dysregulation of PRMTs participates in various human diseases, such as cardiovascular diseases, cancer, diabetes and neurodegenerative disorders. Inflammation is one normal response to infection or injury by immune system, which can keep body homeostasis. Emerging data reveal that inflammation is associated with the development of numerous diseases. Moreover, accumulated evidence proves that PRMTs have been characterized to regulate inflammation in various diseases. In this review article, we delineate the function and molecular mechanism of PRMTs in regulation of inflammation in current literature. Moreover, we discuss that targeting PRMTs by its inhibitors and compounds could have therapeutic potential.
Collapse
Affiliation(s)
- Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jinxin Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Shizhe Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Peter Wang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui 233030, China.
| | - Guanning Shang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
6
|
Shen S, Zhou H, Xiao Z, Zhan S, Tuo Y, Chen D, Pang X, Wang Y, Wang J. PRMT1 in human neoplasm: cancer biology and potential therapeutic target. Cell Commun Signal 2024; 22:102. [PMID: 38326807 PMCID: PMC10851560 DOI: 10.1186/s12964-024-01506-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024] Open
Abstract
Protein arginine methyltransferase 1 (PRMT1), the predominant type I protein arginine methyltransferase, plays a crucial role in normal biological functions by catalyzing the methylation of arginine side chains, specifically monomethylarginine (MMA) and asymmetric dimethylarginine (ADMA), within proteins. Recent investigations have unveiled an association between dysregulated PRMT1 expression and the initiation and progression of tumors, significantly impacting patient prognosis, attributed to PRMT1's involvement in regulating various facets of tumor cell biology, including DNA damage repair, transcriptional and translational regulation, as well as signal transduction. In this review, we present an overview of recent advancements in PRMT1 research across different tumor types, with a specific focus on its contributions to tumor cell proliferation, metastasis, invasion, and drug resistance. Additionally, we expound on the dynamic functions of PRMT1 during distinct stages of cancer progression, elucidating its unique regulatory mechanisms within the same signaling pathway and distinguishing between its promotive and inhibitory effects. Importantly, we sought to provide a comprehensive summary and analysis of recent research progress on PRMT1 in tumors, contributing to a deeper understanding of its role in tumorigenesis, development, and potential treatment strategies.
Collapse
Affiliation(s)
- Shiquan Shen
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Honglong Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zongyu Xiao
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215124, China
| | - Shaofen Zhan
- Department of Neurology, Guangdong Second Provincial General Hospital, Southern Medical University, Guangzhou, 510317, China
| | - Yonghua Tuo
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Danmin Chen
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xiao Pang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yezhong Wang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| | - Ji Wang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
7
|
Feng G, Chen C, Luo Y. PRMT1 accelerates cell proliferation, migration, and tumor growth by upregulating ZEB1/H4R3me2as in thyroid carcinoma. Oncol Rep 2023; 50:210. [PMID: 37859611 PMCID: PMC10603553 DOI: 10.3892/or.2023.8647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
Thyroid carcinoma (TC) represents the most prevalent malignancy of the endocrine system. Protein arginine methyltransferase 1 (PRMT1) is a critical member of the protein arginine methyltransferase family in mammals and is involved in multiple biological processes. This study aimed to investigate the function of PRMT1 in TC. In the present study, human TC cell lines (8505C, CAL62, and BCPAP) and a normal human thyroid cell line Nthy‑ori 3‑1 were employed. Small interfering RNA targeting PRMT1 was used to knock down PRMT1 expression in 8505C cells, and PRMT1 overexpression plasmids were transfected into BCPAP cells. Cell viability was assessed using a CCK‑8 and colony formation assays. Apoptosis was measured using flow cytometry and TUNEL assays. Cell migration was assessed using wound healing and Transwell assays. Reverse transcription‑quantitative PCR was used to determine the mRNA expression levels of PRMT1. Western blotting was used to detect the protein expression levels of PRMT1, E‑cadherin, vimentin, H4R3me2as, and zinc‑finger E homeobox‑binding 1 (ZEB1). Notably, PRMT1 expression was elevated in TC (P<0.01). PRMT1 knockdown inhibited TC cell proliferation and migration and concurrently enhanced migration. Furthermore, PRMT1 knockdown suppressed tumor growth and metastasis in a mouse model of TC. PRMT1 downregulation increased E‑cadherin expression and decreased the expression of vimentin, H4R3me2as, and ZEB1 in the TC cells and the mouse model of TC. Conversely, PRMT1 overexpression had the opposite effect on TC malignant characteristics (P<0.05). These findings suggest that PRMT1 knockdown inhibited TC malignancy by downregulating H4R3me2as/ZEB1, thereby highlighting novel therapeutic targets and diagnostic markers for the management of TC.
Collapse
Affiliation(s)
- Guoli Feng
- Department of General Surgery, Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Changju Chen
- Department of Medical, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yi Luo
- Department of General Surgery, Department of Thyroid and Breast Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
8
|
Zhao X, Sun Y, Xu Z, Cai L, Hu Y, Wang H. Targeting PRMT1 prevents acute and chronic graft-versus-host disease. Mol Ther 2023; 31:3259-3276. [PMID: 37735873 PMCID: PMC10638063 DOI: 10.1016/j.ymthe.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/15/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
Graft-versus-host disease (GVHD) is a common complication after allogeneic hematopoietic stem cell transplantation. Recent studies have reported that protein arginine methyltransferase 1 (PRMT1) is essential for the differentiation and proliferation of T and B cells. Therefore, it is possible that PRMT1 may play a critical role in GVHD. In this study, we observed that PRMT1 expression was upregulated in CD4+ T and B cells from chronic GVHD (cGVHD) patients and mice. However, the prophylactic use of a PRMT1 inhibitor significantly prevented cGVHD in mice by reducing the percentage of T helper (Th)17 cells, germinal center B cells, and plasma cells. The PRMT1 inhibitor also controlled acute GVHD (aGVHD) in mice by decreasing the percentage of Th17 cells. Moreover, inhibiting PRMT1 also weakened Th17 cell differentiation, B cell proliferation, and antibody production in cells from cGVHD patients. Additionally, further studies revealed that PRMT1 regulated B cell proliferation and antibody secretion by methylating isocitrate dehydrogenase 2 (IDH2). We observed asymmetric di-methylation of IDH2 by PRMT1 at arginine 353 promoted IDH2 homodimerization, which enhanced IDH2 activity, further increasing B cell proliferation and antibody production. Collectively, this study provides a rationale for the application of PRMT1 inhibitors in the prevention of aGVHD and cGVHD.
Collapse
Affiliation(s)
- Xiaoyan Zhao
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Sun
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ziwei Xu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Cai
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Huafang Wang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
9
|
Zhao Z, Zhang J, Ren Y, Dong L, Wu H, Hong W, Huang H, Yang X, Pang Z, Wang H. Discovery of 2,4-diphenyl-substituted thiazole derivatives as PRMT1 inhibitors and investigation of their anti-cervical cancer effects. Bioorg Med Chem 2023; 92:117436. [PMID: 37556911 DOI: 10.1016/j.bmc.2023.117436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/11/2023]
Abstract
Cervical cancer is one of the most common cancers that affects middle-aged women and the discovery of new drugs to aid clinical management is needed. As an important member of the protein arginine methyltransferases (PRMTs) family, PRMT1 catalyzes the methylation of protein arginine, which can influence multiple biological processes of cancer cells, such as activating epithelial-mesenchymal transformation (EMT) and acquiring resistance to apoptosis. Therefore, PRMT1 can be considered as a potential drug target for cervical cancer. In the current study, a new sub-binding pocket was discovered by molecular modeling, and by introducing a third substitute on the thiazole group to occupy this pocket, a series of compounds were designed and synthesized as potential PRMT1 inhibitors. Of these, two compounds (ZJG51 and ZJG58) exhibited significant inhibitory activities against PRMT1 without significantly inhibiting PRMT5. Both ZJG51 and ZJG58 displayed potent inhibitory effects on the proliferation of four cancer-derived cell lines and ZJG51 exerted relative selectivity against the cervical cancer cell line, HeLa. Further studies showed that ZJG51 inhibited migration and induce the apoptosis of HeLa cells. Mechanistically, ZJG51 significantly regulated PRMT1 related proteins, and indicated that the induction of apoptosis and inhibition of migration by ZJG51 may involve the activation of Caspase 9 and the inhibition of EMT, respectively. Molecular dynamic simulation and free energy calculation showed that ZJG51 can bind to PRMT1 stably and the binding mode was predicted. These data indicated that introducing the third substitute on the five-membered ring could be a future direction for structure-based optimization of PRMT1 inhibitors, and ZJG51 could be an important lead compound to inform the design of more potent inhibitors.
Collapse
Affiliation(s)
- Ziqi Zhao
- School of Pharmacy, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine (MinZu University of China), Ministry of Education, Beijing 100081, PR China
| | - Jungan Zhang
- School of Pharmacy, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine (MinZu University of China), Ministry of Education, Beijing 100081, PR China
| | - Yixin Ren
- Institute of National Security, Minzu University of China, Beijing 100081, PR China
| | - Luyao Dong
- Beijing Key Laboratory of Antimicrobial Agents/Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Han Wu
- School of Pharmacy, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine (MinZu University of China), Ministry of Education, Beijing 100081, PR China
| | - Wei Hong
- Jingjinji National Center of Technology Innovation, Beijing 100094, PR China
| | - Huoqiang Huang
- School of Pharmacy, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine (MinZu University of China), Ministry of Education, Beijing 100081, PR China
| | - Xinyi Yang
- Beijing Key Laboratory of Antimicrobial Agents/Laboratory of Pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Zongran Pang
- School of Pharmacy, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine (MinZu University of China), Ministry of Education, Beijing 100081, PR China.
| | - Hao Wang
- School of Pharmacy, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine (MinZu University of China), Ministry of Education, Beijing 100081, PR China; Institute of National Security, Minzu University of China, Beijing 100081, PR China.
| |
Collapse
|
10
|
Shin CY, Jang J, Lee HP, Park SH, Kry M, Keo O, Lee BH, Choi W, Lee S, Cho JY. Anti-Oxidative and Anti-Aging Effects of Ethanol Extract of the Officinal Breynia ( Breynia vitis-idaea) In Vitro. PLANTS (BASEL, SWITZERLAND) 2023; 12:1088. [PMID: 36903948 PMCID: PMC10005016 DOI: 10.3390/plants12051088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The skin is the largest organ of the human body, and it is also the one most exposed to external environmental contaminants. The skin is the body's first defense against harmful environmental stimuli, including ultraviolet B (UVB) rays and hazardous chemicals. Therefore, proper care of the skin is required to prevent skin-related diseases and age-related symptoms. In this study, we analyzed anti-aging and anti-oxidative effects of Breynia vitis-idaea ethanol extract (Bv-EE) in human keratinocytes and dermal fibroblasts. The Bv-EE had free radical scavenging activity and decreased the mRNA expression of MMPs and COX-2 in H2O2- or UVB-treated HaCaT cells. The Bv-EE also inhibited AP-1 transcriptional activity and phosphorylation of c-Jun N-terminal kinase, extracellular signal-regulated kinase, and mitogen-activated protein kinase 14 (p38), which are major AP-1 activators upon H2O2 or UVB exposure. Furthermore, the promoter activity and mRNA expression of collagen type I (Col1A1) increased in HDF cells treated with Bv-EE, and Bv-EE recovered the collagen mRNA expression decreased by H2O2 or UVB exposure. These results suggest that Bv-EE has anti-oxidative effects by inhibiting the AP-1 signaling pathway, and shows anti-aging effects by upregulating collagen synthesis.
Collapse
Affiliation(s)
- Chae Yun Shin
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jiwon Jang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hwa Pyoung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Masphal Kry
- Forestry Administration of the Ministry of Agriculture, Phnom Penh 120206, Cambodia
| | - Omaliss Keo
- Forestry Administration of the Ministry of Agriculture, Phnom Penh 120206, Cambodia
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Republic of Korea
| | - Wooram Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Republic of Korea
| | - Jae Youl Cho
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
11
|
Zhang X, Li Z, Shen C, He J, Wang L, Di L, Rui B, Li N, Liu Z. Tao-Hong-Si-Wu decoction improves depressive symptoms in model rats via amelioration of BDNF-CREB-arginase I axis disorders. PHARMACEUTICAL BIOLOGY 2022; 60:1739-1750. [PMID: 36089851 PMCID: PMC9467594 DOI: 10.1080/13880209.2022.2116460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT The traditional Chinese medicine formula Tao-Hong-Si-Wu decoction (TSD), used for treating ischaemic stroke, has the potential to treat depressive disorder (DD). OBJECTIVE To explore the effective targets of TSD on DD animal models. MATERIALS AND METHODS Sprague-Dawley (SD) rats were modelled by inducing chronic unpredictable mild stress (CUMS) during 35 days and treated with three dosages of TSD (2.5, 5 and 10 g/kg) or fluoxetine (10 mg/kg) by oral gavage for 14 days. Bodyweight measurements and behavioural tests were performed to observe the effect of TSD on the CUMS animals. A gas chromatography coupled with mass spectrometry (GC-MS)-based metabolomic analysis was conducted to reveal the metabolic characteristics related to the curative effect of TSD. Levels of the proteins associated with the feature metabolites were analysed. RESULTS Reduced immobile duration and crossed squares in the behavioural tests were raised by 48.6% and 32.9%, on average, respectively, by TSD treatment (ED50=3.2 g/kg). Antidepressant effects of TSD were associated with 13 decreased metabolites and the restorations of ornithine and urea in the serum. TSD (5 g/kg) raised serum serotonin by 54.1 mg/dL but suppressed arginase I (Arg I) by 47.8 mg/dL in the CUMS rats. Proteins on the brain-derived neurotrophic factor (BDNF)-cAMP response element-binding protein (CREB) axis that modulate the inhibition of Arg I were suppressed in the CUMS rats but reversed by the TSD intervention. DISCUSSION AND CONCLUSIONS TSD improves depression-like symptoms in CUMS rats. Further study will focus on the antidepressant-like effects of effective compounds contained in TSD.
Collapse
Affiliation(s)
- Xiaoping Zhang
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Zeng Li
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Chuanpu Shen
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Jinzhi He
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Longfei Wang
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Lei Di
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Bin Rui
- School of Life Science, Anhui Agriculture University, Hefei, China
| | - Ning Li
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| | - Zhicheng Liu
- School of Pharmacy, Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Aizawa Y, Mori M, Suzuki T, Saito A, Inoue H. Shotgun proteomic investigation of methyltransferase and methylation profiles in lipopolysaccharide stimulated RAW264.7 murine macrophages. Biomed Res 2022; 43:73-80. [PMID: 35718447 DOI: 10.2220/biomedres.43.73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Arginine methylation is a common post-translational modification which functions as an epigenetic regulator of transcription and plays a key role in various cell signaling pathways. The methylation of arginine residues is catalyzed by protein arginine methyltransferase (PRMT). However, the expression pattern and underlying mechanism of PRMTs and protein methylation profile in lipopolysaccharide (LPS)-induced innate immune responses are poorly understood. Using a shotgun proteomic approach, we found that LPS stimulation increased arginine and proline metabolism and responses to inflammation and bacterial infections. In comparison, cysteine and methionine metabolism, the pentose phosphate pathway, purine metabolism, and protein methylation factors were also decreased in LPS stimulated murine macrophage cell lines. We revealed that LPS stimulation downregulated PRMT1, PRMT5, and protein arginine methylation profiles in RAW264.7 cells using western blot analysis. Additionally, this phenomenon occurred in parallel with nitric oxide accumulation in LPS-induced macrophages. Using inflammation models, we demonstrate for the first time that LPS stimulation decreases PRMTs, leading to the decreasing of arginine methylation in macrophages.
Collapse
Affiliation(s)
- Yumi Aizawa
- Faculty of Applied Bioscience, Tokyo University of Agriculture.,Research and Development Center for Minimally Invasive Therapies, Institute of Medical Science, Tokyo Medical University
| | - Masaru Mori
- Institute for Advanced Biosciences, Keio University
| | - Tsukasa Suzuki
- Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Akihiro Saito
- Faculty of Applied Bioscience, Tokyo University of Agriculture
| | - Hirofumi Inoue
- Faculty of Applied Bioscience, Tokyo University of Agriculture
| |
Collapse
|
13
|
Shen Q, Xiao Y, Cheng B, Sun Z, Hu Y, Yang H, Luo Y. PRMT1 promotes extracellular matrix degradation and apoptosis of chondrocytes in temporomandibular joint osteoarthritis via the AKT/FOXO1 signaling pathway. Int J Biochem Cell Biol 2021; 141:106112. [PMID: 34715362 DOI: 10.1016/j.biocel.2021.106112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/28/2021] [Accepted: 10/24/2021] [Indexed: 12/16/2022]
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a chronic degenerative joint disease characterized by extracellular matrix (ECM) degradation and chondrocyte apoptosis. The aim of this study was to investigate the role of PRMT1 in TMJOA pathogenesis and its underlying molecular mechanism. Compared to the control group, PRMT1 was highly expressed in IL-1β-treated chondrocytes and articular cartilage following MIA injection into rat TMJs. Furthermore, knocking down PRMT1 considerably inhibited ECM degradation and apoptosis induced by IL-1β. Mechanistic analyses further revealed that PRMT1 knockdown activated the PI3K/AKT signaling pathway and prevented FOXO1 from translocating to the nucleus. Moreover, an inhibitor of AKT (LY294002) rescued the effect of PRMT1 knockdown on IL-1β-induced ECM degradation and apoptosis, and AMI-1, a selective inhibitor of PRMT1, inhibited PRMT1 expression and reversed the pathological progress of TMJOA. Thus, our findings suggest that PRMT1 plays an essential role in ECM degradation and chondrocyte apoptosis in TMJOA via the AKT/FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Qinhao Shen
- Department of Periodontics, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming 650031, Yunnan Province, China; Yunnan Key Laboratory of Stomatology, Kunming 650500, Yunnan Province, China; The First Dental Clinic of the Affiliated Stomatology Hospital of Kunming Medical University, Kunming 650221, Yunnan Province, China
| | - Yiwen Xiao
- Yunnan Key Laboratory of Stomatology, Kunming 650500, Yunnan Province, China; Department of Orthodontics, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming 650031, Yunnan Province, China
| | - Bei Cheng
- Department of Periodontics, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming 650031, Yunnan Province, China; Yunnan Key Laboratory of Stomatology, Kunming 650500, Yunnan Province, China
| | - Zheyi Sun
- Yunnan Key Laboratory of Stomatology, Kunming 650500, Yunnan Province, China
| | - Yu Hu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming 650031, Yunnan Province, China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, Kunming 650500, Yunnan Province, China.
| | - Yingwei Luo
- Department of Periodontics, The Affiliated Stomatological Hospital of Kunming Medical University, Kunming 650031, Yunnan Province, China; The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China.
| |
Collapse
|
14
|
Wang W, Zhong GZ, Long KB, Liu Y, Liu YQ, Xu AL. Silencing miR-181b-5p upregulates PIAS1 to repress oxidative stress and inflammatory response in rats with alcoholic fatty liver disease through inhibiting PRMT1. Int Immunopharmacol 2021; 101:108151. [PMID: 34836796 DOI: 10.1016/j.intimp.2021.108151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE This study aimed to probe the function of microRNA-181b-5p (miR-181b-5p)/protein inhibitor of activated STAT1 (PIAS1)/protein arginine methyltransferase 1 (PRMT1) axis in the progression of alcoholic fatty liver disease (AFLD). METHODS A rat model of AFLD was established and treated with altered miR-181b-5p, PIAS1 or PRMT1 expression constructs to identify their effects on liver function, serum inflammation, liver tissue oxidative stress, hepatocyte apoptosis and pathological changes of liver tissue in rats using a series of assays. miR-181b-5p, PIAS1 and PRMT1 levels were detected, and the targeting relationship between miR-181b-5p and PIAS1 was confirmed. RESULTS MiR-181b-5p and PRMT1 were elevated while PIAS1 was reduced in AFLD rat liver tissues, miR-181b-5p inhibition, PIAS1 overexpression or PRMT1 inhibition improved liver function, attenuated inflammation, oxidative stress, pathological changes and hepatocyte apoptosis in AFLD rat liver tissues. The impacts of miR-181b-5p inhibition on AFLD rats were reversed by PIAS1 silencing. PIAS1 was confirmed as a target gene of miR-181b-5p, and miR-181b-5p regulated PRMT1 expression through binding to PIAS1. CONCLUSION Inhibiting miR-181b-5p can promote the expression of PIAS1, thereby inhibiting PRMT1 and ultimately improving AFLD.
Collapse
Affiliation(s)
- Wei Wang
- Gastroenterology Department, Hunan Aerospace Hospital, Changsha 410205, Hunan, China
| | - Guan-Zhen Zhong
- Gastroenterology Department, Hunan Aerospace Hospital, Changsha 410205, Hunan, China
| | - Kai-Bing Long
- Gastroenterology Department, Hunan Aerospace Hospital, Changsha 410205, Hunan, China
| | - Yang Liu
- Gastroenterology Department, Hunan Aerospace Hospital, Changsha 410205, Hunan, China
| | - Ya-Qian Liu
- Gastroenterology Department, Hunan Aerospace Hospital, Changsha 410205, Hunan, China
| | - Ai-Lei Xu
- Gastroenterology Department, Hunan Aerospace Hospital, Changsha 410205, Hunan, China.
| |
Collapse
|
15
|
Caragana rosea Turcz Methanol Extract Inhibits Lipopolysaccharide-Induced Inflammatory Responses by Suppressing the TLR4/NF-κB/IRF3 Signaling Pathways. Molecules 2021; 26:molecules26216660. [PMID: 34771068 PMCID: PMC8586996 DOI: 10.3390/molecules26216660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 01/05/2023] Open
Abstract
Caragana rosea Turcz, which belongs to the Leguminosae family, is a small shrub found in Northern and Eastern China that is known to possess anti-inflammatory properties and is used to treat fever, asthma, and cough. However, the underlying molecular mechanisms of its anti-inflammatory effects are unknown. Therefore, we used lipopolysaccharide (LPS) in RAW264.7 macrophages to investigate the molecular mechanisms that underlie the anti-inflammatory activities of a methanol extract of Caragana rosea (Cr-ME). We showed that Cr-ME reduced the production of nitric oxide (NO) and mRNA levels of iNOS, TNF-α, and IL-6 in a concentration-dependent manner. We also found that Cr-ME blocked MyD88- and TBK1-induced NF-κB and IRF3 promoter activity, suggesting that it affects multiple targets. Moreover, Cr-ME reduced the phosphorylation levels of IκBα, IKKα/β and IRF3 in a time-dependent manner and regulated the upstream NF-κB proteins Syk and Src, and the IRF3 protein TBK1. Upon overexpression of Src and TBK1, Cr-ME stimulation attenuated the phosphorylation of the NF-κB subunits p50 and p65 and IRF3 signaling. Together, our results suggest that the anti-inflammatory activity of Cr-ME occurs by inhibiting the NF-κB and IRF3 signaling pathways.
Collapse
|
16
|
Ha AT, Kim MY, Cho JY. TAK1/AP-1-Targeted Anti-Inflammatory Effects of Barringtonia augusta Methanol Extract. Molecules 2021; 26:molecules26103053. [PMID: 34065429 PMCID: PMC8160894 DOI: 10.3390/molecules26103053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Barringtonia augusta methanol extract (Ba-ME) is a folk medicine found in the wetlands of Thailand that acts through an anti-inflammatory mechanism that is not understood fully. Here, we examine how the methanol extract of Barringtonia augusta (B. augusta) can suppress the activator protein 1 (AP-1) signaling pathway and study the activities of Ba-ME in the lipopolysaccharide (LPS)-treated RAW264.7 macrophage cell line and an LPS-induced peritonitis mouse model. Non-toxic concentrations of Ba-ME downregulated the mRNA expression of cytokines, such as cyclooxygenase and chemokine ligand 12, in LPS-stimulated RAW264.7 cells. Transfection experiments with the AP-1-Luc construct, HEK293T cells, and luciferase assays were used to assess whether Ba-ME suppressed the AP-1 functional activation. A Western blot assay confirmed that C-Jun N-terminal kinase is a direct pharmacological target of Ba-ME action. The anti-inflammatory effect of Ba-ME, which functions by β-activated kinase 1 (TAK1) inhibition, was confirmed by using an overexpression strategy and a cellular thermal shift assay. In vivo experiments in a mouse model of LPS-induced peritonitis showed the anti-inflammatory effect of Ba-ME on LPS-stimulated macrophages and acute inflammatory mouse models. We conclude that Ba-ME is a promising anti-inflammatory drug targeting TAK1 in the AP-1 pathway.
Collapse
Affiliation(s)
- Anh Thu Ha
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
17
|
Fan F, Podar K. The Role of AP-1 Transcription Factors in Plasma Cell Biology and Multiple Myeloma Pathophysiology. Cancers (Basel) 2021; 13:2326. [PMID: 34066181 PMCID: PMC8151277 DOI: 10.3390/cancers13102326] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy characterized by the clonal expansion of malignant plasma cells within the bone marrow. Activator Protein-1 (AP-1) transcription factors (TFs), comprised of the JUN, FOS, ATF and MAF multigene families, are implicated in a plethora of physiologic processes and tumorigenesis including plasma cell differentiation and MM pathogenesis. Depending on the genetic background, the tumor stage, and cues of the tumor microenvironment, specific dimeric AP-1 complexes are formed. For example, AP-1 complexes containing Fra-1, Fra-2 and B-ATF play central roles in the transcriptional control of B cell development and plasma cell differentiation, while dysregulation of AP-1 family members c-Maf, c-Jun, and JunB is associated with MM cell proliferation, survival, drug resistance, bone marrow angiogenesis, and bone disease. The present review article summarizes our up-to-date knowledge on the role of AP-1 family members in plasma cell differentiation and MM pathophysiology. Moreover, it discusses novel, rationally derived approaches to therapeutically target AP-1 TFs, including protein-protein and protein-DNA binding inhibitors, epigenetic modifiers and natural products.
Collapse
Affiliation(s)
- Fengjuan Fan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China;
| | - Klaus Podar
- Department of Internal Medicine II, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr.-Karl-Dorrek-Strasse 30, 3500 Krems an der Donau, Austria
| |
Collapse
|
18
|
DNA or Protein Methylation-Dependent Regulation of Activator Protein-1 Function. Cells 2021; 10:cells10020461. [PMID: 33670008 PMCID: PMC7926996 DOI: 10.3390/cells10020461] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Epigenetic regulation and modification govern the transcriptional mechanisms that promote disease initiation and progression, but can also control the oncogenic processes, cell signaling networks, immunogenicity, and immune cells involved in anti-inflammatory and anti-tumor responses. The study of epigenetic mechanisms could have important implications for the development of potential anti-inflammatory treatments and anti-cancer immunotherapies. In this review, we have described the key role of epigenetic progression: DNA methylation, histone methylation or modification, and protein methylation, with an emphasis on the activator protein-1 (AP-1) signaling pathway. Transcription factor AP-1 regulates multiple genes and is involved in diverse cellular processes, including survival, differentiation, apoptosis, and development. Here, the AP-1 regulatory mechanism by DNA, histone, or protein methylation was also reviewed. Various methyltransferases activate or suppress AP-1 activities in diverse ways. We summarize the current studies on epigenetic alterations, which regulate AP-1 signaling during inflammation, cancer, and autoimmune diseases, and discuss the epigenetic mechanisms involved in the regulation of AP-1 signaling.
Collapse
|
19
|
Bednarz-Misa I, Fortuna P, Fleszar MG, Lewandowski Ł, Diakowska D, Rosińczuk J, Krzystek-Korpacka M. Esophageal Squamous Cell Carcinoma Is Accompanied by Local and Systemic Changes in L-arginine/NO Pathway. Int J Mol Sci 2020; 21:E6282. [PMID: 32872669 PMCID: PMC7503331 DOI: 10.3390/ijms21176282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
The L-arginine/NO pathway holds promise as a source of potential therapy target and biomarker; yet, its status and utility in esophageal squamous cell carcinoma (ESCC) is unclear. We aimed at quantifying pathway metabolites in sera from patients with ESCC (n = 61) and benign conditions (n = 62) using LC-QTOF-MS and enzyme expression in esophageal tumors and matched noncancerous samples (n = 40) using real-time PCR with reference to ESCC pathology and circulating immune/inflammatory mediators, quantified using Luminex xMAP technology. ESCC was associated with elevated systemic arginine and asymmetric dimethylarginine. Citrulline decreased and arginine bioavailability increased along with increasing ESCC advancement. Compared to adjacent tissue, tumors overexpressed ODC1, NOS2, PRMT1, and PRMT5 but had downregulated ARG1, ARG2, and DDAH1. Except for markedly higher NOS2 and lower ODC1 in tumors from M1 patients, the pathology-associated changes in enzyme expression were subtle and present also in noncancerous tissue. Both the local enzyme expression level and systemic metabolite concentration were related to circulating inflammatory and immune mediators, particularly those associated with eosinophils and those promoting viability and self-renewal of cancer stem cells. Metabolic reprogramming in ESCC manifests itself by the altered L-arginine/NO pathway. Upregulation of PRMTs in addition to NOS2 and ODC1 and the pathway link with stemness-promoting cytokines warrants further investigation.
Collapse
Affiliation(s)
- Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (M.G.F.); (Ł.L.)
| | - Paulina Fortuna
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (M.G.F.); (Ł.L.)
| | - Mariusz G. Fleszar
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (M.G.F.); (Ł.L.)
| | - Łukasz Lewandowski
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (M.G.F.); (Ł.L.)
| | - Dorota Diakowska
- Department of Gastrointestinal and General Surgery, Wroclaw Medical University, 50-368 Wroclaw, Poland;
- Department of Nervous System Diseases, Wroclaw Medical University, 51-618 Wroclaw, Poland;
| | - Joanna Rosińczuk
- Department of Nervous System Diseases, Wroclaw Medical University, 51-618 Wroclaw, Poland;
| | - Małgorzata Krzystek-Korpacka
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (M.G.F.); (Ł.L.)
| |
Collapse
|