1
|
Yu X, Wang X, Xu F, Zhang X, Wang M, Zhou R, Sun Z, Pan X, Feng L, Zhang W, Sun Y, Zhang W, Zhou D, Jiang Y. Mir-615-3p promotes osteosarcoma progression via the SESN2/AMPK/mTOR pathway. Cancer Cell Int 2024; 24:411. [PMID: 39702297 DOI: 10.1186/s12935-024-03604-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary malignant bone neoplasm. Growing researches have highlighted the tumor promoting role of miR-615-3p in various cancers. Notwithstanding, the biological function and underlying mechanisms of miR-615-3p in OS development still unclear. METHODS Quantitative Real-Time PCR analysis (qRT-PCR) and RNA fluorescence in situ hybridization (FISH) staining were performed to measure miR-615-3p expression in OS. CCK-8 assay, colony formation assay and EdU assay were applied to analyze the OS cell proliferation activity. Cell metastasis abilities were evaluated using Transwell assays. Analysis of apoptosis was performed based on flow cytometric detection. The potential mechanisms of miR-615-3p in OS progression were investigated through RNA immunoprecipitation (RIP) assays, dual-luciferase reporter assays, qRT-PCR and western blotting. In vivo experiments, mouse xenograft model was carried out to assess the tumorigenicity of miR-615-3p. RESULTS This study demonstrated a significant upregulation of miR-615-3p in OS. In addition, miR-615-3p knockdown suppressed OS proliferation, invasion, metastasis and EMT. Mechanistically, miR-615-3p regulated sestrin 2 (SESN2) expression negatively by targeting its 3'UTR. Moreover, silencing SESN2 facilitated OS progression and activated mTOR pathway. Noteworthy, the anticancer functions of miR-615-3p knockdown were partially recovered by SESN2 silencing. Taken together, the miR-615-3p/SESN2/mTOR pathway is critical for regulating OS progression. CONCLUSION Our results revealed that miR-615-3p modulated mTOR signaling, thus influencing the progression of OS. For OS treatment, molecular strategies that target the miR-615-3p/SESN2/mTOR pathway is promising.
Collapse
Affiliation(s)
- Xuecheng Yu
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Xin Wang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Fan Xu
- Department of Disease Control, 987 Hospital of Joint Logistics Support Force of PLA, Baoji, Shaanxi, China
| | - Xinyi Zhang
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Muyi Wang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Ruikai Zhou
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Zhengyi Sun
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China
| | - Xiaohui Pan
- Department of Orthopedics, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu Province, China
| | - Lin Feng
- The people's hospital of WuQia county, WuQia, 845450, Xinjiang, China
| | - Wanchao Zhang
- Department of Radiology, The people's hospital of WuQia county, WuQia, 845450, Xinjiang, China
| | - Yong Sun
- Department of Orthopedics, Wuqia People's Hospital, Xinjiang, China
| | - Wenting Zhang
- Affiliated Changzhou Children's Hospital of Nantong University, Changzhou, 213003, Jiangsu, China.
| | - Dong Zhou
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China.
- Department of Orthopedics, Wuqia People's Hospital, Xinjiang, China.
- Affiliated Changzhou Children's Hospital of Nantong University, Changzhou, 213003, Jiangsu, China.
- Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China.
| | - Yuqing Jiang
- Department of Orthopedics, Changzhou Medical Center, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Nanjing Medical University, Changzhou, 213003, China.
| |
Collapse
|
2
|
Shen J, Lai Y, Wu Y, Lin X, Zhang C, Liu H. Ubiquitination in osteosarcoma: unveiling the impact on cell biology and therapeutic strategies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0231. [PMID: 39475222 PMCID: PMC11523277 DOI: 10.20892/j.issn.2095-3941.2024.0231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/30/2024] [Indexed: 11/02/2024] Open
Abstract
Ubiquitination, a multifaceted post-translational modification, regulates protein function, degradation, and gene expression. The pivotal role of ubiquitination in the pathogenesis and progression of cancer, including colorectal, breast, and liver cancer, is well-established. Osteosarcoma, an aggressive bone tumor predominantly affecting adolescents, also exhibits dysregulation of the ubiquitination system, encompassing both ubiquitination and deubiquitination processes. This dysregulation is now recognized as a key driver of osteosarcoma development, progression, and chemoresistance. This review highlights recent progress in elucidating how ubiquitination modulates tumor behavior across signaling pathways. We then focus on the mechanisms by which ubiquitination influences osteosarcoma cell function. Finally, we discuss the potential for targeting the ubiquitin-proteasome system in osteosarcoma therapy. By unraveling the impact of ubiquitination on osteosarcoma cell physiology, we aim to facilitate the development of novel strategies for prognosis, staging, treatment, and overcoming chemoresistance.
Collapse
Affiliation(s)
- Jianlin Shen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian 351100, China
- Central Laboratory, Affiliated Hospital of Putian University, Putian 351100, China
| | - Yue Lai
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yanjiao Wu
- Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan 528000, China
| | - Xuan Lin
- Department of Environmental and Biological Engineering, Putian University, Putian 351100, China
| | - Cheng Zhang
- Department of Trauma Center, Zhongda Hospital, Southeast University, Nanjing 210000, China
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
3
|
Colaco JC, Suresh B, Kaushal K, Singh V, Ramakrishna S. The Role of Deubiquitinating Enzymes in Primary Bone Cancer. Mol Biotechnol 2024:10.1007/s12033-024-01254-y. [PMID: 39177860 DOI: 10.1007/s12033-024-01254-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/01/2024] [Indexed: 08/24/2024]
Abstract
Bone is a living, intricate, and dynamic tissue providing locomotion and protection of the body. It also performs hematopoiesis and mineral homeostasis. Osteosarcoma (OS), Ewing sarcoma (ES), and chondrosarcoma (CS) are primary bone cancers. OS and ES mostly develop in younger individuals, and CS generally develops in adults. Ubiquitination regulates numerous cellular processes. The deubiquitinating enzymes (DUBs) detach the ubiquitin molecules from the ubiquitin labeled substrate, altering ubiquitinated protein functions and regulating protein stability via various signaling pathways. Protein homeostasis and bone remodeling are both crucially influenced by the UPS. Recently, there have been several reports on DUBs involved in bone homeostasis and various bone disorders through the regulation of osteoblasts and osteoclasts via NF-κB, Wnt/β-catenin, TRAF6, TGFβ, ERK1/2, and PI3K/Akt pathways. However, DUBs regulating function in bone homeostasis is still in its infancy. Here, we summarized several recent identifications on DUBs, with a focus on their role in bone cancer progression. Therefore, the study attempts to summarize association with the expression level of DUBs as key factors driving bone cancers and also provide new insights on DUBs as key pharmacologic targets for bone cancer therapeutics.
Collapse
Affiliation(s)
- Jencia Carminha Colaco
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Kamini Kaushal
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, 382715, India.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea.
- College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
4
|
Mao P, Feng Z, Liu Y, Zhang K, Zhao G, Lei Z, Di T, Zhang H. The Role of Ubiquitination in Osteosarcoma Development and Therapies. Biomolecules 2024; 14:791. [PMID: 39062505 PMCID: PMC11274928 DOI: 10.3390/biom14070791] [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: 04/15/2024] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The ubiquitin-proteasome system (UPS) maintains intracellular protein homeostasis and cellular function by regulating various biological processes. Ubiquitination, a common post-translational modification, plays a crucial role in the regulation of protein degradation, signal transduction, and other physiological and pathological processes, and is involved in the pathogenesis of various cancers, including osteosarcoma. Osteosarcoma, the most common primary malignant bone tumor, is characterized by high metastatic potential and poor prognosis. It is a refractory bone disease, and the main treatment modalities are surgery combined with chemotherapy. Increasing evidence suggests a close association between UPS abnormalities and the progression of osteosarcoma. Due to the complexity and pleiotropy of the ubiquitination system, each step in the ubiquitination process can be targeted by drugs. In recent years, research and development of inhibitors targeting the ubiquitin system have increased gradually, showing great potential for clinical application. This article reviews the role of the ubiquitination system in the development and treatment of osteosarcoma, as well as research progress, with the hope of improving the therapeutic effects and prognosis of osteosarcoma patients by targeting effective molecules in the ubiquitination system.
Collapse
Affiliation(s)
- Peng Mao
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopaedics of Gansu Province, Lanzhou University, Lanzhou 730030, China
| | - Zuxi Feng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Yong Liu
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopaedics of Gansu Province, Lanzhou University, Lanzhou 730030, China
| | - Kai Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopaedics of Gansu Province, Lanzhou University, Lanzhou 730030, China
| | - Guanghai Zhao
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopaedics of Gansu Province, Lanzhou University, Lanzhou 730030, China
| | - Zeyuan Lei
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopaedics of Gansu Province, Lanzhou University, Lanzhou 730030, China
| | - Tianning Di
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
| | - Haihong Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Second Clinical School, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopaedics of Gansu Province, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
5
|
Li M, Dai M, Cheng B, Li S, Guo E, Fu J, Ma T, Yu B. Strategies that regulate LSD1 for novel therapeutics. Acta Pharm Sin B 2024; 14:1494-1507. [PMID: 38572094 PMCID: PMC10985039 DOI: 10.1016/j.apsb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 04/05/2024] Open
Abstract
Histone methylation plays crucial roles in regulating chromatin structure and gene transcription in epigenetic modifications. Lysine-specific demethylase 1 (LSD1), the first identified histone demethylase, is universally overexpressed in various diseases. LSD1 dysregulation is closely associated with cancer, viral infections, and neurodegenerative diseases, etc., making it a promising therapeutic target. Several LSD1 inhibitors and two small-molecule degraders (UM171 and BEA-17) have entered the clinical stage. LSD1 can remove methyl groups from histone 3 at lysine 4 or lysine 9 (H3K4 or H3K9), resulting in either transcription repression or activation. While the roles of LSD1 in transcriptional regulation are well-established, studies have revealed that LSD1 can also be dynamically regulated by other factors. For example, the expression or activity of LSD1 can be regulated by many proteins that form transcriptional corepressor complexes with LSD1. Moreover, some post-transcriptional modifications and cellular metabolites can also regulate LSD1 expression or its demethylase activity. Therefore, in this review, we will systematically summarize how proteins involved in the transcriptional corepressor complex, various post-translational modifications, and metabolites act as regulatory factors for LSD1 activity.
Collapse
Affiliation(s)
- Meng Li
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Mengge Dai
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Bing Cheng
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Shaotong Li
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Enhui Guo
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Junwei Fu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
| | - Ting Ma
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Henan Normal University, Xinxiang 453007, China
| | - Bin Yu
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450000, China
- Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Henan Normal University, Xinxiang 453007, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
6
|
Wang T, Huang J, Chen G, Fu J, Li T, Zou X, Yi H. miR-1293 suppresses osteosarcoma progression by modulating drug sensitivity in response to cisplatin treatment. Int Immunopharmacol 2024; 130:111702. [PMID: 38367464 DOI: 10.1016/j.intimp.2024.111702] [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: 01/17/2024] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
Chemotherapy is considered the primary treatment for osteosarcoma. however, its effectiveness is limited due to drug resistance and toxicity. Thus, identifying novel therapeutic targets to enhance the efficacy of chemotherapy is urgently needed. Here, we identified a novel cisplatin-sensitivity enhancing mechanism via up-regulation of the tumour suppressor gene, miR-1293. Meanwhile, higher levels of miR-1293 observed in prechemotherapy patients were associated with a more favorable prognosis. The mechanism underlying cisplatin upregulated miR-1293 expression involves hypomethylation of the miR-1293 promoter, which blocks the binding of the transcription repressor TFAP2A to the promoter. Furthermore, miR-1293 inhibits osteosarcoma progression by targeting TIMP1 to inactivate the Notch1/Hes1 and TGFBR1/Smad2/3 pathways, thereby promoting tumour cell death. The findings presented herein unveil a novel mechanism for enhancing cisplatin sensitivity and proposed a potential therapeutic strategy for osteosarcoma through pre-chemotherapy supplementation of miR-1293.
Collapse
Affiliation(s)
- Tingxuan Wang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510005, China
| | - Jincheng Huang
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450000, China.
| | - Gang Chen
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Jiahui Fu
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou 510005, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 10032, China.
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510005, China.
| | - Hualin Yi
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510005, China; Guangzhou National Laboratory, Guangzhou 510005, China.
| |
Collapse
|
7
|
Li A, Wang S, Nie J, Xiao S, Xie X, Zhang Y, Tong W, Yao G, Liu N, Dan F, Shu Z, Liu J, Liu Z, Yang F. USP3 promotes osteosarcoma progression via deubiquitinating EPHA2 and activating the PI3K/AKT signaling pathway. Cell Death Dis 2024; 15:235. [PMID: 38531846 PMCID: PMC10965993 DOI: 10.1038/s41419-024-06624-7] [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: 10/02/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
Ubiquitin-specific protease 3 (USP3) plays an important role in the progression of various tumors. However, the role of USP3 in osteosarcoma (OS) remains poorly understood. The aim of this study was to explore the biological function of USP3 in OS and the underlying molecular mechanism. We found that OS had higher USP3 expression compared with that of normal bone tissue, and high expression of USP3 was associated with poor prognosis in patients with OS. Overexpression of USP3 significantly increased OS cell proliferation, migration, and invasion. Mechanistically, USP3 led to the activation of the PI3K/AKT signaling pathway in OS by binding to EPHA2 and then reducing its protein degradation. Notably, the truncation mutant USP3-F2 (159-520) interacted with EPHA2, and amino acid 203 was found to play an important role in this process. And knockdown of EPHA2 expression reversed the pro-tumour effects of USP3-upregulating. Thus, our study indicates the USP3/EPHA2 axis may be a novel potential target for OS treatment.
Collapse
Affiliation(s)
- Anan Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shijiang Wang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiangbo Nie
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shining Xiao
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinsheng Xie
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yu Zhang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Weilai Tong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Geliang Yao
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ning Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Dan
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhiguo Shu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiaming Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhili Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Feng Yang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Institute of Spine and Spinal Cord, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
8
|
Han X, Yin M, Gong C, Zhang C, Zhu G, Hu M, Tan K, Jiang L, Wang G, Li L. A1BG-AS1 promotes the biological functions of osteosarcoma cells via regulating the microRNA-148a-3p/USP22 axis and stabilizing the expression of SIRT1 through deubiquitinase function. Expert Opin Ther Targets 2023; 27:1017-1029. [PMID: 37747800 DOI: 10.1080/14728222.2023.2263908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 09/24/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND The study aims to explore the role of A1BG antisense RNA 1 (A1BG-AS1), microRNA (miR)-148a-3p and ubiquitin-specific protease 22 (USP22) on osteosarcoma (OS) cell growth. RESEARCH DESIGN & METHODS A1BG-AS1, miR-148a-3p, USP22, and silent information regulator 2 homolog 1 (SIRT1) levels in OS tissues and cells were determined. The effects of A1BG-AS1, miR-148a-3p, and USP22 on the biological functions of OS cells were examined by functional assays. In vivo assay was conducted to observe the effect of A1BG-AS1 on OS growth in vitro. The relationship of A1BG-AS1, miR-148a-3p, and USP22 was analyzed by bioinformatics analysis, RNA-fluorescence in situ hybridization, luciferase activity, and RNA binding protein immunoprecipitation assays. The relation between USP22 and SIRT1 was evaluated by immunoprecipitation. RESULTS A1BG-AS1 and USP22 were highly expressed, and miR-148a-3p was lowly expressed in OS tissues and cells. Down-regulation of A1BG-AS1 and USP22 or up-regulation of miR-148a-3p impaired the malignant behaviors of OS cells. A1BG-AS1 sponged miR-148a-3p, and miR-148a-3p targeted USP22, thereby inhibiting USP22 expression. Up-regulating USP22 reversed the A1BG-AS1 suppression-induced phenotypic inhibition of OS cells. USP22 affected the biological functions of OS cells by deubiquitinating SIRT1. CONCLUSION A1BG-AS1 facilitates the biological functions of OS cells via mediating the miR-148a-3p/USP22 axis.
Collapse
Affiliation(s)
- Xiuxin Han
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Mengfan Yin
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
- Department of Orthopedic Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Chen Gong
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Genbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Mengxue Hu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - La Jiang
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, Anhui, China
| |
Collapse
|
9
|
Luo W, Zhang G, Wang Z, Wu Y, Xiong Y. Ubiquitin-specific proteases: Vital regulatory molecules in bone and bone-related diseases. Int Immunopharmacol 2023; 118:110075. [PMID: 36989900 DOI: 10.1016/j.intimp.2023.110075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/06/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023]
Abstract
Stabilization of bone structure and function involves multiple cell-to-cell and molecular interactions, in which the regulatory functions of post-translational modifications such as ubiquitination and deubiquitination shouldn't be underestimated. As the largest family of deubiquitinating enzymes, the ubiquitin-specific proteases (USPs) participate in the development of bone homeostasis and bone-related diseases through multiple classical osteogenic and osteolytic signaling pathways, such as BMP/TGF-β pathway, NF-κB/p65 pathway, EGFR-MAPK pathway and Wnt/β-catenin pathway. Meanwhile, USPs may also broadly regulate regulate hormone expression level, cell proliferation and differentiation, and may further influence bone homeostasis from gene fusion and nuclear translocation of transcription factors. The number of patients with bone-related diseases is currently enormous, making exploration of their pathogenesis and targeted therapy a hot topic. Pathological increases in the levels of inflammatory mediators such as IL-1β and TNF-α lead to inflammatory bone diseases such as osteoarthritis, rheumatoid arthritis and periodontitis. While impaired body metabolism greatly increases the probability of osteoporosis. Abnormal physiological activity of bone-associated cells results in a variety of bone tumors. The regulatory role of USPs in bone-related disease has received particular attention from academics in recent studies. In this review, we focuse on the roles and mechanisms of USPs in bone homeostasis and bone-related diseases, with the expectation of informing targeted therapies in the clinic.
Collapse
Affiliation(s)
- Wenxin Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guorui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhanqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yingying Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Xu J, Yang G, An W, Wang W, Li F, Meng Y, Wang X. Correlations between the severity of radiation-induced oral mucositis and salivary epidermal growth factor as well as inflammatory cytokines in patients with head and neck cancer. Head Neck 2023; 45:1122-1129. [PMID: 36866530 DOI: 10.1002/hed.27313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/29/2023] [Accepted: 02/08/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND This study aimed to investigate correlations of the severity of radiation-induced oral mucositis (RIOM) with epidermal growth factor (EGF) and inflammatory cytokines in patients with head and neck cancer (HNC). METHODS Levels of inflammatory cytokines and EGF in saliva of HNC patients were measured. Correlations of inflammatory cytokines and EGF levels with RIOM severity and pain degree, and their diagnostic values on RIOM severity were determined. RESULTS Elevated IFN-γ, TNF-α, IL-2, and IL-6 levels, and reduced IL-4, IL-10, and EGF levels were found in patients with severe RIOM. IFN-γ, TNF-α, IL-2, and IL-6 were positively correlated with RIOM severity, while IL-10, IL-4, and EGF were negatively correlated with it. All factors were effective in predicting the severity of RIOM. CONCLUSION IFN-γ, TNF-α, IL-2, and IL-6 in saliva of patients with HNC are positively correlated with the severity of RIOM, and IL-4, IL-10, and EGF were negatively correlated with it.
Collapse
Affiliation(s)
- Juan Xu
- Department of Head and Neck Radiotherapy Ward 1, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Guang Yang
- Department of Head and Neck Radiotherapy Ward 1, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei An
- Department of Head and Neck Radiotherapy Ward 1, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei Wang
- Department of Head and Neck Radiotherapy Ward 1, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fangfang Li
- Department of Head and Neck Radiotherapy Ward 1, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yingtao Meng
- Department of Nursing, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shangdong, China
| | - Xingli Wang
- Department of Nursing, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shangdong, China
| |
Collapse
|
11
|
Dreher RD, Theisen ER. Lysine specific demethylase 1 is a molecular driver and therapeutic target in sarcoma. Front Oncol 2023; 12:1076581. [PMID: 36686841 PMCID: PMC9846348 DOI: 10.3389/fonc.2022.1076581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/07/2022] [Indexed: 01/05/2023] Open
Abstract
Sarcomas are a diverse group of tumors with numerous oncogenic drivers, and display varied clinical behaviors and prognoses. This complexity makes diagnosis and the development of new and effective treatments challenging. An incomplete understanding of both cell of origin and the biological drivers of sarcomas complicates efforts to develop clinically relevant model systems and find new molecular targets. Notably, the histone lysine specific demethylase 1 (LSD1) is overexpressed in a number of different sarcomas and is a potential therapeutic target in these malignancies. With the ability to modify histone marks, LSD1 is a key player in many protein complexes that epigenetically regulate gene expression. It is a largely context dependent enzyme, having vastly different and often opposing roles depending on the cellular environment and which interaction partners are involved. LSD1 has been implicated in the development of many different types of cancer, but its role in bone and soft tissue sarcomas remains poorly understood. In this review, we compiled what is known about the LSD1 function in various sarcomas, to determine where knowledge is lacking and to find what theme emerge to characterize how LSD1 is a key molecular driver in bone and soft tissue sarcoma. We further discuss the current clinical landscape for the development of LSD1 inhibitors and where sarcomas have been included in early clinical trials.
Collapse
Affiliation(s)
- Rachel D. Dreher
- Abigail Wexner Research Institute, Center for Childhood Cancer and Blood Diseases, Nationwide Children’s Hospital, Columbus, OH, United States
- Biomedical Sciences Graduate Program, College of Medicine, the Ohio State University, Columbus, OH, United States
| | - Emily R. Theisen
- Abigail Wexner Research Institute, Center for Childhood Cancer and Blood Diseases, Nationwide Children’s Hospital, Columbus, OH, United States
- Biomedical Sciences Graduate Program, College of Medicine, the Ohio State University, Columbus, OH, United States
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
12
|
Liu X, Wang M, Zhang L, Huang L. LncRNA ZFAS1 contributes to osteosarcoma progression via miR-520b and miR-520e-mediated inhibition of RHOC signaling. Clinics (Sao Paulo) 2023; 78:100143. [PMID: 36473367 PMCID: PMC9727593 DOI: 10.1016/j.clinsp.2022.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES We examined the expression of Lnc-ZFAS1 in osteosarcoma and comprehensively evaluated its effects on osteosarcoma in vitro and vivo. Moreover, we revealed the regulatory mechanism between Lnc-ZFAS1 and miR-520b/miR-520e-mediated RHOC and provided a novel clue for ameliorating osteosarcoma. METHOD The expression of Long non-coding RNA Zinc Finger Antisense 1 (LncRNA ZFAS1) osteosarcoma tissues and normal tissues in the TCGA database was analyzed. Then, LncRNA ZFAS1 expression was further verified in clinical samples and osteosarcoma cell lines (U2OS and KHOS), as well as the human osteoblast cell line hFOB1.19 by qRT-PCR. Thereafter, LncRNA ZFAS1 was overexpressed or silenced to explore its effects on cell proliferation, apoptosis, migration, invasion, and Epithelial-Mesenchymal Transition (EMT). The fundamental mechanism through which Lnc-ZFAS1 affects osteosarcoma progression was further investigated and verified. RESULTS We found that LncRNA ZFAS1 was upregulated in osteosarcoma, and Lnc-ZFAS1 overexpression facilitated osteosarcoma cells proliferation, migration, invasion and EMT, while Lnc-ZFAS1 silence exerted reverse influence. Mechanistically, Lnc-ZFAS1 functionally acted as a sponger of microRNA-520b (miR-520b) and microRNA-520e (miR-520e) to up-regulate Ras Homologue C (RHOC). In addition, depleted Lnc-ZFAS1 restrained osteosarcoma cells proliferation, migration, and invasion, which could be rescued by RHOC overexpression. Lnc-ZFAS1 was upregulated in osteosarcoma and Lnc-ZFAS1 could exert promoted impact upon osteosarcoma cells proliferation, migration, invasion, and EMT in vitro. CONCLUSIONS Lnc-ZFAS1 acted sponger of miR-520b and miR-520e to promote RHOC, indicating that Lnc-ZFAS1/miR-520b/RHOC and Lnc-ZFAS1/miR-520e/RHOC axes might serve as potential therapeutic strategies against osteosarcoma.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China
| | - Mingyang Wang
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China
| | - Liwen Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China
| | - Lei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China.
| |
Collapse
|
13
|
Jin S, Kudo Y, Horiguchi T. The Role of Deubiquitinating Enzyme in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2022; 24:ijms24010552. [PMID: 36613989 PMCID: PMC9820089 DOI: 10.3390/ijms24010552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
Ubiquitination and deubiquitination are two popular ways for the post-translational modification of proteins. These two modifications affect intracellular localization, stability, and function of target proteins. The process of deubiquitination is involved in histone modification, cell cycle regulation, cell differentiation, apoptosis, endocytosis, autophagy, and DNA repair after damage. Moreover, it is involved in the processes of carcinogenesis and cancer development. In this review, we discuss these issues in understanding deubiquitinating enzyme (DUB) function in head and neck squamous cell carcinoma (HNSCC), and their potential therapeutic strategies for HNSCC patients are also discussed.
Collapse
|
14
|
Song J, Yuan X, Piao L, Wang J, Wang P, Zhuang M, Liu J, Liu Z. Cellular functions and molecular mechanisms of ubiquitination in osteosarcoma. Front Oncol 2022; 12:1072701. [PMID: 36530999 PMCID: PMC9753703 DOI: 10.3389/fonc.2022.1072701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/17/2022] [Indexed: 02/13/2025] Open
Abstract
Although some advances have been made in the treatment of osteosarcoma in recent years, surgical resection remains the mainstream treatment. Initial and early diagnosis of osteosarcoma could be very difficult to achieve due to the insufficient sensitivity for the means of examination. The distal metastasis of osteosarcoma also predicts the poor prognosis of osteosarcoma. In order to solve this series of problems, people begin to discover a new method of diagnosing and treating osteosarcoma. Ubiquitination, as an emerging posttranslational modification, has been shown to be closely related to osteosarcoma in studies over the past decades. In general, this review describes the cellular functions and molecular mechanisms of ubiquitination during the development of osteosarcoma.
Collapse
Affiliation(s)
- Jiaxun Song
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaofeng Yuan
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lianhua Piao
- Jiangsu University of Technology, Changzhou, China
| | - Jiawen Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Pu Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ming Zhuang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jie Liu
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhiwei Liu
- The Third Affiliated Hospital of Soochow University, Changzhou, China
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
15
|
Chen T, Liu J, Zhang H, Li J, Shang G. Long Intergenic Noncoding RNA00265 Enhances Cell Viability and Metastasis via Targeting miR-485-5p/USP22 Axis in Osteosarcoma. Front Oncol 2022; 12:907472. [PMID: 35692754 PMCID: PMC9179024 DOI: 10.3389/fonc.2022.907472] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/19/2022] [Indexed: 01/09/2023] Open
Abstract
Osteosarcoma is one of the bone malignancies in children and adolescents. Long noncoding RNAs (lncRNAs) have been demonstrated to participate in osteosarcoma development and progression. Linc00265 has been shown to involve in osteosarcoma oncogenesis; however, the underlying mechanism is largely unclear. In this study, we investigated the function of linc00265 in osteosarcoma cells, including cell viability, migration and invasion. Moreover, we elucidated mechanistically the involvement of linc00265 in osteosarcoma. We found that linc00265 overexpression promoted viability, migration and invasion of osteosarcoma cells. Notably, linc00265 sponged miR-485-5p and increased the expression of USP22, one target of miR-485-5p, in osteosarcoma cells. Strikingly, linc00265 exerted its oncogenic function via regulating miR-485-5p and USP22 in osteosarcoma. Taken together, targeting linc00265 is a promising approach for treating osteosarcoma patients.
Collapse
|
16
|
Yuan B, Liu H, Pan X, Dong X, Qu LF, Sun J, Pan LL. LSD1 downregulates p21 expression in vascular smooth muscle cells and promotes neointima formation. Biochem Pharmacol 2022; 198:114947. [PMID: 35143753 DOI: 10.1016/j.bcp.2022.114947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 01/10/2023]
Abstract
Neointima formation is characterized by the proliferation of vascular smooth muscle cells (VSMC). Although lysine-specific demethylase 1 (LSD1) has critical functions in several diseases, its role in neointima formation remains to be clarified. In this study, we aimed to explore the crucial role of LSD1 on neointima formation using a carotid artery injury model in mice. We observed that aberrant LSD1 expression was increased in human and mouse stenotic arteries and platelet-derived growth factor-BB (PDGF-BB)-treated VSMC. Furthermore, LSD1 knockdown significantly mitigated neointima formation in vivo and inhibited PDGF-BB-induced VSMC proliferation in vitro. We further uncovered that LSD1 overexpression exhibited opposite phenotypes in vivo and in vitro. Finally, LSD1 knockdown inhibited VSMC proliferation by increasing p21 expression, which is associated with LSD1 mediated di-methylated histone H3 on lysine 4 (H3K4me2) modification. Taken together, our data suggest that LSD1 may be a potential therapeutic target for the treatment of neointima formation.
Collapse
Affiliation(s)
- Baohui Yuan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - He Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoliang Dong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Le-Feng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China; School of Food Science and Technology, Jiangnan University, Wuxi, China.
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
17
|
Zhang P, Xu K, Wang J, Zhang J, Quan H. Identification of N6-methylandenosine related LncRNAs biomarkers associated with the overall survival of osteosarcoma. BMC Cancer 2021; 21:1285. [PMID: 34852770 PMCID: PMC8638368 DOI: 10.1186/s12885-021-09011-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Osteosarcoma (OS) is a differentiation disease caused by the genetic and epigenetic differentiation of mesenchymal stem cells into osteoblasts. OS is a common, highly malignant tumor in children and adolescents. Fifteen to 20 % of the patients find distant metastases at their first visit. The purpose of our study was to identify biomarkers for tracking the prognosis and treatment of OS to improve the survival rate of patients. MATERIALS AND METHODS In this study, which was based on Therapeutically Applicable Research to Generate Effective Treatments (TARGET), we searched for m6A related lncRNAs in OS. We constructed a network between lncRNA and m6A, and built an OS prognostic risk model. RESULTS We identified 14,581 lncRNAs by using the dataset from TARGET. We obtained 111 m6A-related lncRNAs through a Pearson correlation analysis. A network was built between lncRNA and m6A genes. Eight m6A-related lncRNAs associated with survival were identified through a univariate Cox analysis. A selection operator (LASSO) Cox regression was used to construct a prognostic risk model with six genes (RP11-286E11.1, LINC01426, AC010127.3, DLGAP1-AS2, RP4-657D16.3, AC002398.11) obtained through least absolute shrinkage. We also discovered upregulated levels of DLGAP1-AS2 and m6A methylation in osteosarcoma tissues/cells compared with normal tissues/osteoblasts cells. CONCLUSION We constructed a risk score prognosis model of m6A-related lncRNAs (RP11-286E11.1, LINC01426, AC010127.3, DLGAP1-AS2, RP4-657D16.3, AC002398.11) using the dataset downloaded from TRAGET. We verified the value of the model by dividing all samples into test groups and training groups. However, the role of m6A-related lncRNAs in osteosarcoma needs to be further researched by cell and in vivo studies.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Keteng Xu
- Department of Joint surgery, Huangshan City People's Hospital, Huangshan, Anhui, China.
| | - Jingcheng Wang
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China. .,Department of Orthopedics, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, China.
| | - Jiale Zhang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Huahong Quan
- Department of Graduate, Dalian Medical University, Dalian, 116044, Liaoning, China
| |
Collapse
|
18
|
Mullard M, Lavaud M, Regnier L, Tesfaye R, Ory B, Rédini F, Verrecchia F. Ubiquitin-specific proteases as therapeutic targets in paediatric primary bone tumours? Biochem Pharmacol 2021; 194:114797. [PMID: 34678225 DOI: 10.1016/j.bcp.2021.114797] [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: 09/01/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/04/2023]
Abstract
In children and young adults, primary malignant bone tumours are mainly composed of osteosarcoma and Ewing's sarcoma. Despite advances in treatments, nearly 40% of patients succumb to these diseases. In particular, the clinical outcome of metastatic osteosarcoma or Ewing's sarcoma remains poor, with less than 30% of patients who develop metastases surviving five years after initial diagnosis. Over the last decade, the cancer research community has shown considerable interest in the processes of protein ubiquitination and deubiquitination. In particular, a growing number of studies show the relevance to target the ubiquitin-specific protease (USP) family in various cancers. This review provides an update on the current knowledge regarding the implication of these USPs in the progression of bone sarcoma: osteosarcoma and Ewing's sarcoma.
Collapse
Affiliation(s)
- Mathilde Mullard
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France
| | - Mélanie Lavaud
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France
| | - Laura Regnier
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France
| | - Robel Tesfaye
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France
| | - Benjamin Ory
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France
| | - Françoise Rédini
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France
| | - Franck Verrecchia
- INSERM, Université de Nantes, UMR1238, "Bone Sarcoma and Remodelling of Calcified Tissues", 44000 Nantes, France.
| |
Collapse
|
19
|
Feng T, Ling S, Xu C, Ying L, Su D, Xu X. Ubiquitin-specific peptidase 22 in cancer. Cancer Lett 2021; 514:30-37. [PMID: 33989708 DOI: 10.1016/j.canlet.2021.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/21/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023]
Abstract
Recently, many studies have shown that deubiquitination modification of proteins is of great significance in major physiological processes such as cell proliferation, apoptosis, and differentiation. The ubiquitin-specific peptidase (USP) family is one of the most numerous and structurally diverse of the deubiquitinates known to date. USP22, an important member of the USP family, has been found to be closely associated with tumor cell cycle regulation, stemness maintenance, invasion and metastasis, chemoresistance, and immune regulation. We focus on recent advances regarding USP22's function in cancer and discuss the prospect of USP22 in this review.
Collapse
Affiliation(s)
- Tingting Feng
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Colorectal Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Sunbin Ling
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chenyang Xu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Lisha Ying
- Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Dan Su
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|