1
|
Wang Y, Wu Z, Wang C, Wu N, Wang C, Hu S, Shi J. The role of WWP1 and WWP2 in bone/cartilage development and diseases. Mol Cell Biochem 2024; 479:2907-2919. [PMID: 38252355 DOI: 10.1007/s11010-023-04917-7] [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: 08/18/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024]
Abstract
Bone and cartilage diseases are often associated with trauma and senescence, manifested as pain and limited mobility. The repair of bone and cartilage lesion by mesenchymal stem cells is regulated by various transcription factors. WW domain-containing protein 1 (WWP1) and WW domain-containing protein 2 (WWP2) are named for WW domain which recognizes PPXY (phono Ser Pro and Pro Arg) motifs of substrate. WWP1and WWP2 are prominent components of the homologous to the E6-AP carboxyl terminus (HECT) subfamily, a group of the ubiquitin ligase. Recently, some studies have found that WWP1 and WWP2 play an important role in the pathogenesis of bone and cartilage diseases and regulate the level and the transactivation of various transcription factors through ubiquitination. Therefore, this review summarizes the distribution and effects of WWP1 and WWP2 in the development of bone and cartilage, discusses the potential mechanism and therapeutic drugs in bone and cartilage diseases such as osteoarthritis, fracture, and osteoporosis.
Collapse
Affiliation(s)
- Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Cunyi Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Na Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Chenyu Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Shiyu Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China
| | - Jiejun Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
2
|
Yao W, Kulyar MFEA, Ding Y, Du H, Hong J, Loon KS, Nawaz S, Li J. The Effect of miR-140-5p with HDAC4 towards Growth and Differentiation Signaling of Chondrocytes in Thiram-Induced Tibial Dyschondroplasia. Int J Mol Sci 2023; 24:10975. [PMID: 37446153 DOI: 10.3390/ijms241310975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
There is evidence to suggest that microRNA-140-5p (miR-140), which acts as a suppressor, is often elevated and has a role in various malignancies. Nevertheless, neither the function nor the mechanisms in chondrocytes linked with bone disorders, e.g., tibial dyschondroplasia (TD), have been satisfactorily established. The purpose of this study was to look into the role of microRNA-140-5p (miR-140) and its interaction with HDAC4 in chondrocytes, as well as the implications for tibial dyschondroplasia (TD), with a particular focus on the relationship between low miR-140 expression and poor pathologic characteristics, as well as its physiological effects on chondrocyte growth, differentiation, and chondrodysplasia. In this investigation, we discovered that TD had a reduced expression level of the miR-140. There was a correlation between low miR-140 expression, poor pathologic characteristics, and the short overall survival of chondrocytes. Our findings show an aberrant reduction in miR-140 expression, and HDAC4 overexpression caused disengagement in resting and proliferation zones. This further resulted in uncontrolled cell proliferation, differentiation, and chondrodysplasia. Mechanistically, HDAC4 inhibited the downstream transcription factors MEF2C and Runx2 and interacted with Col-Ⅱ, Col-X, and COMP. However, miR-140 binding to the 3'-UTR of HDAC4 resulted in the growth and differentiation of chondrocytes. Moreover, the expression of HDAC4 through LMK-235 was significantly decreased, and the expression was significantly increased under ITSA-1, referring to a positive feedback circuit of miR-140 and HDAC4 for endochondral bone ossification. Furthermore, as a prospective treatment, the flavonoids of Rhizoma drynariae (TFRD) therapy increased the expression of miR-140. Compared to the TD group, TFRD treatment increased the expression of growth-promoting and chondrocyte differentiation markers, implying that TFRD can promote chondrocyte proliferation and differentiation in the tibial growth plate. Hence, directing this circuit may represent a promising target for chondrocyte-related bone disorders and all associated pathological bone conditions.
Collapse
Affiliation(s)
- Wangyuan Yao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA 92521, USA
| | - Muhammad Fakhar-E-Alam Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yanmei Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Haitao Du
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiajia Hong
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Kyein San Loon
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
3
|
Wang K, Wang N, Liu J, Zhou J, Lei S, Yue H, Feng H, Feng K, Kang X. Silencing circular RNA hsa_circABCC1 inhibits osteosarcoma progression through down-regulating HDAC4 via sponging miR-591. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37052456 DOI: 10.1002/tox.23786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/18/2023] [Accepted: 03/12/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Circular RNA (circRNA) has been shown to play an important regulatory role in the development of various cancers, including osteosarcoma (OS). However, the role of circRNA ABCC1 (circABCC1) in OS was still poorly understood. The aim of our study was to investigate the role of circABCC1 in OS progression and its potential molecular mechanisms. METHODS The expression of circABCC1, microRNA-591 (miR-591) and histone deacetylase 4 (HDAC4) in OS tissues or cells was detected by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot (WB) analyses. In vitro experiments, the viability, proliferation, apoptosis, migration, invasion and autophagy of U2OS and HOS cells were assessed in vitro using cell counting kit-8 (CCK-8) assay, 5-ethynyl-29-deoxyuridine (EdU) assay, flow cytometry (FCM) assay, transwell migration and invasion assays (transwell) and WB assay, respectively. Interactions between circABCC1 and miR-591, miR-591 and HDAC4 were confirmed using a dual luciferase reporter gene assay system. The oncogenic role of circABCC1 in OS in vivo was examined by establishing a tumor xenograft model. RESULTS CircABCC1 was significantly elevated in OS tissues (about 3.1-folds) and cells (U2OS (about 2.1-folds) and HOS (about 2.8-folds)) compared with the control (p < .05). Silencing of circABCC1 significantly reduced the viability and proliferation, promoted apoptosis, impaired migration and invasion, and increased autophagy of U2OS and HOS cells (p < .05). In addition, miR-591 was confirmed to be a target of circABCC1, exerting an opposite effect to circABCC1 (p < .05). MiR-591 attenuation in U2OS and HOS cells was able to reply to the inhibition of cell proliferation, migration and invasion as well as promotion of cell apoptosis and autophagy mediated by silencing circABCC1 (p < .05). HDAC4 was verified to be the target gene of miR-591 in U2OS and HOS cells and was regulated by the circABCC1/miR-591 axis (p < .05), and restoration of HDAC4 levels in U2OS and HOS cells was able to restore the altered cellular function caused by silencing circABCC1 (p < .05). In addition, knockdown of circABCC1 attenuated tumor growth in vivo (p < .05). CONCLUSION Silencing of circABCC1 inhibits osteosarcoma progression by attenuating HDAC4 expression through sponging miR-591.
Collapse
Affiliation(s)
- Kai Wang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Na Wang
- Department of Oncology, Second Hospital of Lanzhou University, Lanzhou, China
| | - Jingsheng Liu
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Jianwei Zhou
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Shuanhu Lei
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Haiyuan Yue
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Haijun Feng
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Kai Feng
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| | - Xuewen Kang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Yu Y, Dong G, Li Z, Zheng Y, Shi Z, Wang G. circ‑LRP6 contributes to osteosarcoma progression by regulating the miR‑141‑3p/HDAC4/HMGB1 axis. Int J Oncol 2022; 60:38. [PMID: 35211755 PMCID: PMC8878724 DOI: 10.3892/ijo.2022.5328] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
Circular RNA-lipoprotein receptor 6 (circ-LRP6) serves a role in promoting the tumorigenesis of retinoblastoma, esophageal squamous cell cancer and oral squamous cell carcinoma; however, whether circ-LRP6 demonstrates the same effect in osteosarcoma (OS) is yet to be fully elucidated. The present study aimed to analyze the expression, role and potential molecular mechanism of circ-LRP6 in OS. The expression levels of circ-LRP6, microRNA (miR)-141-3p, histone deacetylase 4 (HDAC4) and high mobility group protein 1 (HMGB1) were evaluated by reverse transcription- quantitative PCR in OS tissues and cell lines. Cell Counting Kit-8, Transwell and Matrigel assays were conducted to evaluate cell proliferation, migration and invasion, respectively. Western blotting was also performed to determine HDAC4 and HMGB1 protein expression levels. Bioinformatics and dual-luciferase reporter assays were used to predict and analyze the interactions between circ-LRP6 and miR-141-3p, miR-141-3p and HDAC4, as well as between miR-141-3p and HMGB1. Additionally, RNA immunoprecipitation was performed to verify the association between circ-LRP6 and miR-141-3p. The results confirmed that circ-LRP6 was highly expressed in OS tissues and cell lines. In addition, circ-LRP6 negatively regulated the expression of miR-141-3p and, in turn, miR-141-3p negatively regulated HDAC4 and HMGB1 expression. Functional assays revealed that circ-LRP6 knockdown inhibited the proliferation, migration and invasion of OS cells, whereas the inhibition of miR-141-3p or the overexpression of either HDAC4 or HMGB1 partly reversed the inhibitory effect of circ-LRP6 knockdown. In summary, the present study determined that circ-LRP6 knockdown inhibited the proliferation, migration and invasion of OS cells by regulating the miR-141-3p/HDAC4/HMGB1 axis.
Collapse
Affiliation(s)
- Yali Yu
- Department of Laboratory, Zhengzhou Orthopedic Hospital, Zhengzhou, Henan 450052, P.R. China
| | - Guixiang Dong
- Department of Laboratory, Zhengzhou Orthopedic Hospital, Zhengzhou, Henan 450052, P.R. China
| | - Zijun Li
- Department of Laboratory, Zhengzhou Orthopedic Hospital, Zhengzhou, Henan 450052, P.R. China
| | - Yan Zheng
- Department of Laboratory, Zhengzhou Orthopedic Hospital, Zhengzhou, Henan 450052, P.R. China
| | - Zhisong Shi
- Department of Orthopedic Surgery, Zhumadian Central Hospital, Zhumadian, Henan 463000, P.R. China
| | - Guanghui Wang
- Department of Orthopedic Surgery, Zhumadian Central Hospital, Zhumadian, Henan 463000, P.R. China
| |
Collapse
|
5
|
Abstract
Osteosarcoma is the most common primary bone malignancy in adolescents. Its high propensity to metastasize is the leading cause for treatment failure and poor prognosis. Although the research of osteosarcoma has greatly expanded in the past decades, the knowledge and new therapy strategies targeting metastatic progression remain sparse. The prognosis of patients with metastasis is still unsatisfactory. There is resonating urgency for a thorough and deeper understanding of molecular mechanisms underlying osteosarcoma to develop innovative therapies targeting metastasis. Toward the goal of elaborating the characteristics and biological behavior of metastatic osteosarcoma, it is essential to combine the diverse investigations that are performed at molecular, cellular, and animal levels from basic research to clinical translation spanning chemical, physical sciences, and biology. This review focuses on the metastatic process, regulatory networks involving key molecules and signaling pathways, the role of microenvironment, osteoclast, angiogenesis, metabolism, immunity, and noncoding RNAs in osteosarcoma metastasis. The aim of this review is to provide an overview of current research advances, with the hope to discovery druggable targets and promising therapy strategies for osteosarcoma metastasis and thus to overcome this clinical impasse.
Collapse
Affiliation(s)
- Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Liu W, Wang D, Liu L, Wang L, Yan M. miR-140 inhibits osteosarcoma progression by impairing USP22-mediated LSD1 stabilization and promoting p21 expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:436-448. [PMID: 33868787 PMCID: PMC8040122 DOI: 10.1016/j.omtn.2021.01.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 01/22/2021] [Indexed: 12/29/2022]
Abstract
Osteosarcoma is a bone tumor frequently diagnosed in children and young adults. Despite advances in chemotherapy and surgical resection, tumors metastasize in 30% of osteosarcoma patients. In addition, side effects caused by chemotherapeutic drugs, as well as the development of chemoresistance, highlight the need to identify the molecular mechanisms involved in the pathogenesis of osteosarcoma. We compared 65 osteosarcoma samples to their adjacent normal tissues, as well as commercially obtained osteosarcoma cell lines with normal osteoblast cell lines, and identified a role for the microRNA (miR)-140/ubiquitin-specific protease 22 (USP22)/lysine-specific demethylase 1 (LSD1)/p21 axis in the development of osteosarcoma. Osteosarcoma tissues and cells exhibited poor miR-140 and p21 expression, whereas the expression of USP22 and LSD1 was increased. Overexpression of miR-140 inhibited cell proliferation, migration, and invasion and promoted cell apoptosis by directly targeting USP22, resulting in its decreased expression. Overexpression of USP22 reversed the effects of miR-140 overexpression in osteosarcoma cells. Overexpression of miR-140 or USP22 knockdown led to the ubiquitination and degradation of LSD1. miR-140 overexpression also suppressed tumorigenesis in vivo. This study revealed a role for miR-140 in the restriction of osteosarcoma development and identified miR-140 as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Dunwei Wang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Lidi Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Lin Wang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Ming Yan
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China
| |
Collapse
|
7
|
Zhu J, Wang J, Huang J, Du W, He Y, Pan H, Luo J. MicroRNA-140-5p regulates the proliferation, apoptosis and inflammation of RA FLSs by repressing STAT3. Exp Ther Med 2020; 21:171. [PMID: 33456538 PMCID: PMC7792473 DOI: 10.3892/etm.2020.9602] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Ectopic expression of microRNA (miRNA) in rheumatoid arthritis (RA) fibroblast-like synoviocyte (RA FLS) is associated with the development of rheumatoid arthritis. The present study aimed to evaluate the effects of miRNA-140-5p (miR-140) on the properties of RA FLSs. It was found that miR-140 expression was decreased in 33 RA patients and extracted RA FLS samples, when compared to the corresponding healthy controls. Abnormally increased miR-140 expression in RA FLSs attenuated cell proliferation and increased cell apoptosis. Additionally, reduced pro-inflammatory cytokine production was observed in RA FLSs transfected with a miR-140 precursor. Furthermore, the 3'-UTR of the signal transducer and activator of transcription (STAT) 3 gene was identified as a target of miR-140. Notably, restoration of STAT3 expression rescued the regulatory effect of miR-140 on the proliferation, apoptosis and inflammatory cytokine production of RA FLSs. Therefore, the current findings indicated that miR-140 is a crucial modulator of both proliferation and apoptosis, shedding light on the etiology behind RA FLS viability, which is modulated by an interplay between miR-140 and STAT3 in the context of RA.
Collapse
Affiliation(s)
- Jiehua Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianglin Wang
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jialin Huang
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Wensheng Du
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Yingzhong He
- Department of Laboratory Medicine, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hongfei Pan
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Junmin Luo
- Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
8
|
Zhang S, Gong Y, Li C, Yang W, Li L. Beyond regulations at DNA levels: A review of epigenetic therapeutics targeting cancer stem cells. Cell Prolif 2020; 54:e12963. [PMID: 33314500 PMCID: PMC7848960 DOI: 10.1111/cpr.12963] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/05/2023] Open
Abstract
In the past few years, the paramount role of cancer stem cells (CSCs), in terms of cancer initiation, proliferation, metastasis, invasion and chemoresistance, has been revealed by accumulating studies. However, this level of cellular plasticity cannot be entirely explained by genetic mutations. Research on epigenetic modifications as a complementary explanation for the properties of CSCs has been increasing over the past several years. Notably, therapeutic strategies are currently being developed in an effort to reverse aberrant epigenetic alterations using specific chemical inhibitors. In this review, we summarize the current understanding of CSCs and their role in cancer progression, and provide an overview of epigenetic alterations seen in CSCs. Importantly, we focus on primary cancer therapies that target the epigenetic modification of CSCs by the use of specific chemical inhibitors, such as histone deacetylase (HDAC) inhibitors, DNA methyltransferase (DNMT) inhibitors and microRNA‐based (miRNA‐based) therapeutics.
Collapse
Affiliation(s)
- Shunhao Zhang
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Sichuan Province, Chengdu, China
| | - Yanji Gong
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, Department of Temporomandibular Joint, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China.,State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Chunjie Li
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| | - Wenbin Yang
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Sichuan Province, Chengdu, China
| | - Longjiang Li
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral Disease, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
9
|
Ni Y, Yang Y, Ran J, Zhang L, Yao M, Liu Z, Zhang L. miR-15a-5p inhibits metastasis and lipid metabolism by suppressing histone acetylation in lung cancer. Free Radic Biol Med 2020; 161:150-162. [PMID: 33059020 DOI: 10.1016/j.freeradbiomed.2020.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/04/2020] [Accepted: 10/07/2020] [Indexed: 02/05/2023]
Abstract
Metabolic reprogramme was a key characteristic of malignant tumors. Increased evidences indicated that besides Warburg effect (abnormal glucose metabolism), abnormal lipid metabolism played more and more important in progression and metastasis of malignant tumors. MiR-15a-5p could inhibit development of lung cancer, while its regulating mechanism, especially the role in lipid metabolism still remained unclear. In this study, we confirmed that miR-15a-5p inhibited proliferation, migration and invasion of lung cancer cells. The online analysis of Mirpath v.3 predicted that miR-15a-5p was closely associated with fatty acid synthesis and lipid metabolism. In vitro cell experiments revealed that miR-15a-5p significantly suppressed fatty acid synthesis of lung cancer cells by inhibiting acetate uptake. Extensive analysis indicated that miR-15a-5p could suppress acetyl-CoA activity and decrease histone H4 acetylation by inhibiting ACSS2 expression. In addition, we also observed that ACSS2 located in nucleus under hypoxic conditions, while miR-15a-5p could be transported into nucleus to inhibit the function of ACSS2. Our study unveiled a novel mechanism of miR-15a-5p in inhibiting metastasis of lung cancer cells by suppressing lipid metabolism via suppression of ACSS2 mediated acetyl-CoA activity and histone acetylation.
Collapse
Affiliation(s)
- Yinyun Ni
- Precision Medicine Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ying Yang
- Precision Medicine Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jingjing Ran
- Precision Medicine Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Lu Zhang
- West China-Washington Mitochondria and Metabolism Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Menglin Yao
- Precision Medicine Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Zhiqiang Liu
- Precision Medicine Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Li Zhang
- Precision Medicine Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China; Laboratory of Pathology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Liu J, Feng G, Li Z, Li R, Xia P. Knockdown of CircCRIM1 Inhibits HDAC4 to Impede Osteosarcoma Proliferation, Migration, and Invasion and Facilitate Autophagy by Targeting miR-432-5p. Cancer Manag Res 2020; 12:10199-10210. [PMID: 33116874 PMCID: PMC7573324 DOI: 10.2147/cmar.s253130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/08/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) serve for a genre of considerable modulatory molecules that have been largely researched in human cancers. However, the contribution of circRNA cysteine-rich transmembrane bone morphogenetic protein regulator 1 (circCRIM1) to osteosarcoma (OS) is completely unclear. METHODS All the RNA levels were examined via quantitative real-time polymerase chain reaction (qRT-PCR). Cellular proliferation and migration/invasion were, respectively, analyzed using 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide (MTT) assay and transwell assay. The determination of all protein expression was administrated by Western blot. Dual-luciferase reporter assay was used for proving the target combination. The exploration of circCRIM1 in vivo was performed by xenograft assay. RESULTS In OS tissues and cells, circCRIM1 was differentially up-regulated. Functionally, cell proliferation, migration and invasion were suppressed while autophagy was promoted after circCRIM1 was down-regulated in OS cells. Mechanistically, mircoRNA-432-5p (miR-432-5p) was a miRNA target of circCRIM1 and the inhibitory effect of circCRIM1 knockdown on OS progression was achieved by targeting miR-432-5p. Moreover, histone deacetylase 4 (HDAC4) was a downstream gene of miR-432-5p and circCRIM1 targeted miR-432-5p to up-regulate HDAC4 level. MiR-432-5p inhibited proliferation, migration, and invasion but enhanced autophagy of OS cells through down-regulating HDAC4. In vivo, knockdown of circCRIM1 decreased OS growth via acting on the miR-432-5p/HDAC4 axis. CONCLUSION Our findings elucidated the oncogenic function of circCRIM1 in OS via the regulation of the miR-432-5p/HDAC4 axis, affording a novel view about how circRNA participated in OS development.
Collapse
Affiliation(s)
- Jun Liu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin130000, People’s Republic of China
| | - Guang Feng
- The Fourth Medical Center of PLA General Hospital, Beijing100048, People’s Republic of China
| | - Zhengwei Li
- Department of Orthopaedics, The Second Hospital of Jilin University, Jilin130000, People’s Republic of China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin130000, People’s Republic of China
| | - Peng Xia
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin130000, People’s Republic of China
| |
Collapse
|
11
|
Liu W, Liu P, Gao H, Wang X, Yan M. Long non-coding RNA PGM5-AS1 promotes epithelial-mesenchymal transition, invasion and metastasis of osteosarcoma cells by impairing miR-140-5p-mediated FBN1 inhibition. Mol Oncol 2020; 14:2660-2677. [PMID: 32412676 PMCID: PMC7530781 DOI: 10.1002/1878-0261.12711] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/14/2020] [Accepted: 05/11/2020] [Indexed: 01/31/2023] Open
Abstract
Osteosarcoma is an uncommon tumor occurring in bone, accompanied by elevated incidence and reduced rate of healing. Epithelial‐to‐mesenchymal transition (EMT) serves as a conceptual paradigm to explain the invasion and metastasis of osteosarcoma and other cancers. Hence, developing effective therapeutic strategy to treat the EMT of osteosarcoma is essential. Here, we identified the molecular mechanism of long noncoding RNA (lncRNA) PGM5‐AS1 in EMT and progression of osteosarcoma. Microarray‐based analysis was employed to screen the osteosarcoma‐related differentially expressed lncRNAs. The levels of PGM5‐AS1 as well as microRNA‐140‐5p (miR‐140‐5p) and fibrillin‐1 (FBN1) in osteosarcoma tissues and cells were determined. Dual‐luciferase reporter gene assay, RNA pull‐down assay, and RNA immunoprecipitation assay were conducted to validate the relationship among PGM5‐AS1, miR‐140‐5p, and FBN1. Expression of PGM5‐AS1, miR‐140‐5p, and FBN1 was altered by overexpression, shRNA, mimic, or inhibitors in order to investigate how they regulated migration, invasion, and EMT of osteosarcoma cells in vitro. Loss‐ and gain‐of‐function approaches were employed in nude mice to detect their roles in tumorigenesis in vivo. Osteosarcoma tissues and cells exhibited low expression of miR‐140‐5p, but high expression of PGM5‐AS1 and FBN1. PGM5‐AS1 competitively bound to miR‐140‐5p to upregulate FBN1. Furthermore, hindering PGM5‐AS1 and FBN1 or overexpressing miR‐140‐5p dampened migration, invasion, and EMT of osteosarcoma cells in vitro. Furthermore, silencing PGM5‐AS1 or FBN1, or overexpressing miR‐140‐5p markedly inhibited tumorigenesis in nude mice in vivo. Taken together, PGM5‐AS1 depletion causes FBN1 reduction to retard osteosarcoma processes by negatively modulating miR‐140‐5p.
Collapse
Affiliation(s)
- Wei Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Pengcheng Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hang Gao
- Department of Bone and Joint Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ming Yan
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Guo Q, Zhang N, Liu S, Pang Z, Chen Z. By targeting TRAF6, miR-140-3p inhibits TGF-β1-induced human osteosarcoma epithelial-to-mesenchymal transition, migration, and invasion. Biotechnol Lett 2020; 42:2123-2133. [PMID: 32562135 DOI: 10.1007/s10529-020-02943-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/13/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVES We evaluated the effects of miR-140-3p on EMT, cellular migration, and invasion in TGF-β1 treated human OS cells. Human fresh OS tissue and normal bone tissue specimens were gathered from 42 patients (29 male and 13 female, 11 to 24 years of age with a mean age of 17.5 ± 2.3 years) diagnosed with OS by pathology. By targeting TRAF6, miR-140-3p inhibits TGF-β1-induced human osteosarcoma epithelial-to-mesenchymal transition, migration, and invasion. RESULTS In this study, we found microRNA (miR)-140-3p to be down-regulated and tumor necrosis factor receptor-associated factor 6 (TRAF6) to be up-regulated in patient OS samples. Lower levels of miR-140-3p and higher levels of TRAF6 were found in the advanced Enneking stage of OS. Furthermore, both mRNA and protein levels of TRAF6 were negatively associated with miR-140-3p mRNA expression in human OS tissue. TRAF6 was verified as a direct target of miR-140-3p in TGF-β1-treated human U2OS cells. Further, a miR-140-3p mimic dramatically inhibited while a miR-140-3p inhibitor enhanced TGF-β1-induced epithelial-to-mesenchymal transition, migration, and invasion of U2OS cells. Small interfering RNA was found to silence TRAF6 and to partly reverse the effects of the miR-140-3p inhibitor on TGF-β1-treated U2OS cells in vitro. CONCLUSION These results demonstrate miR-140-3p to function as a tumor inhibitor of human OS cells by decreasing TRAF6 expression. miR-140-3p and TRAF6 may be valuable and novel biomarkers for diagnosis and treatment of OS.
Collapse
Affiliation(s)
- Qianchen Guo
- Department of Orthopaedics, General Hospital of Tianjin Medical University, 154, Anshan road, Heping district, Tianjin, 300052, China.
| | - Nai Zhang
- Department of Neurosurgery, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Shen Liu
- Department of Orthopaedics, General Hospital of Tianjin Medical University, 154, Anshan road, Heping district, Tianjin, 300052, China
| | - Zixuan Pang
- Department of Orthopaedics, The Seventh People's Hospital of Hebei Province, Dingzhou, 073000, Hebei, China
| | - Zhao Chen
- Department of Orthopaedics, General Hospital of Tianjin Medical University, 154, Anshan road, Heping district, Tianjin, 300052, China
| |
Collapse
|
13
|
Chen Z, Zhang Z, Guo L, Wei X, Zhang Y, Wang X, Wei L. The role of histone deacetylase 4 during chondrocyte hypertrophy and endochondral bone development. Bone Joint Res 2020; 9:82-89. [PMID: 32435460 PMCID: PMC7229302 DOI: 10.1302/2046-3758.92.bjr-2019-0172.r1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chondrocyte hypertrophy represents a crucial turning point during endochondral bone development. This process is tightly regulated by various factors, constituting a regulatory network that maintains normal bone development. Histone deacetylase 4 (HDAC4) is the most well-characterized member of the HDAC class IIa family and participates in different signalling networks during development in various tissues by promoting chromatin condensation and transcriptional repression. Studies have reported that HDAC4-null mice display premature ossification of developing bones due to ectopic and early-onset chondrocyte hypertrophy. Overexpression of HDAC4 in proliferating chondrocytes inhibits hypertrophy and ossification of developing bones, which suggests that HDAC4, as a negative regulator, is involved in the network regulating chondrocyte hypertrophy. Overall, HDAC4 plays a key role during bone development and disease. Thus, understanding the role of HDAC4 during chondrocyte hypertrophy and endochondral bone formation and its features regarding the structure, function, and regulation of this process will not only provide new insight into the mechanisms by which HDAC4 is involved in chondrocyte hypertrophy and endochondral bone development, but will also create a platform for developing a therapeutic strategy for related diseases. Cite this article:Bone Joint Res. 2020;9(2):82–89.
Collapse
Affiliation(s)
- Zhi Chen
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhiwei Zhang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Li Guo
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaochun Wei
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yang Zhang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojian Wang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wei
- Department of Orthopedics, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
14
|
Wu H, Liu C, Yang Q, Xin C, Du J, Sun F, Zhou L. MIR145-3p promotes autophagy and enhances bortezomib sensitivity in multiple myeloma by targeting HDAC4. Autophagy 2020; 16:683-697. [PMID: 31242129 PMCID: PMC7138223 DOI: 10.1080/15548627.2019.1635380] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 06/19/2019] [Accepted: 06/19/2019] [Indexed: 01/04/2023] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy with poor survival. Autophagy, a stress-responsive catabolic process mediated by lysosomal activity, plays a crucial role in the pathophysiology of MM. Growing evidence has indicated that dysregulated microRNAs (miRNAs) are associated with the aberrant autophagy in various human cancers. However, to date, few miRNAs have been reported to directly modulate autophagy in the pathobiology of MM. In this study, we investigated the role of MIR145-3p (microRNA 145-3p) in MM, with focus on cellular processes autophagy and cell death. Our results provided evidence that downregulation of MIR145-3p expression was associated with disease progression in human MM. MIR145-3p triggered autophagic flux through direct targeting of HDAC4 (histone deacetylase 4) in MM cells, leading to enhanced apoptosis. Silencing HDAC4 recapitulated the effects of MIR145-3p, whereas enforced expression of HDAC4 abrogated the effects of MIR145-3p. Furthermore, we showed that suppression of HDAC4 by MIR145-3p resulted in upregulation of the pro-apoptotic protein BCL2L11 and caused MTORC1 inactivation, which in turn led to enhanced autophagy and cell death. Importantly, we demonstrated that MIR145-3p mimic could potentiate the anti-MM activity of bortezomib in both in vitro and in vivo experiments. Overall, our findings indicate that MIR145-3p exerted a tumor suppression function in MM by inducing autophagic cell death and suggest that MIR145-3p-based targeted therapy would represent a novel strategy for MM treatment.Abbreviations: 3-MA: 3-methyladenine; 3'-UTR: 3'-untranslated region; 7-AAD: 7-aminoactinomycin D; ACTB: actin beta; ANXA5: annexin A5; ATG5: autophagy related 5; ATG7: autophagy related 7; B2M: beta-2-microglobulin; BAF: bafilomycin A1; BCL2L11: BCL2 like 11; Bort: bortezomib; CASP3: caspase 3; CCK-8: Cell Counting Kit-8; CQ: chloroquine; Ct: threshold cycle; ctrl: control; DAPI: 4',6-diamidino-2-phenylindole; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HDAC4: histone deacetylase 4; ISS: International Staging System; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; miRNAs: microRNAs; MIR145-3p: microRNA 145-3p; MM: multiple myeloma; mRNA: messenger RNA; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; PCs: plasma cells; PFS: progression-free survival; qRT-PCR: quantitative reverse transcription PCR; RPS6KB1: ribosomal protein S6 kinase B1; SD: standard deviation; siRNA: small interfering RNA; SQSTM1: sequestosome 1; STV: starvation; TUBB: tubulin beta class I.
Collapse
Affiliation(s)
- Hongkun Wu
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Chang Liu
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Qingyuan Yang
- Department of Clinical Laboratory Medicine, Tenth People’s Hospital of Tongji University, Shanghai, P.R. China
| | - Chengde Xin
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Juan Du
- Department of Hematology, The Myeloma & Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Tenth People’s Hospital of Tongji University, Shanghai, P.R. China
| | - Lin Zhou
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| |
Collapse
|
15
|
Targeting the Cancer Epigenome with Histone Deacetylase Inhibitors in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:55-75. [PMID: 32767234 DOI: 10.1007/978-3-030-43085-6_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epigenetic deregulation is an emerging hallmark of cancer that enables tumor cells to escape surveillance by tumor suppressors and ultimately progress. The structure of the epigenome consists of covalent modifications of chromatin components, including acetylation by histone acetyltransferases (HATs) and deacetylation by histone deacetylases (HDACs). Targeting these enzymes with inhibitors to restore epigenetic homeostasis has been explored for many cancers. Osteosarcoma, an aggressive bone malignancy that primarily affects children and young adults, is notable for widespread genetic and epigenetic instability. This may explain why therapy directed at unique molecular pathways has failed to substantially improve outcomes in osteosarcoma over the past four decades. In this review, we discuss the potential of targeting the cancer epigenome, with a focus on histone deacetylase inhibitors (HDACi) for osteosarcoma. We additionally highlight the safety and tolerance of HDACi, combination chemotherapy with HDACi, and the ongoing challenges in the development of these agents.
Collapse
|
16
|
de Azevedo JWV, de Medeiros Fernandes TAA, Fernandes JV, de Azevedo JCV, Lanza DCF, Bezerra CM, Andrade VS, de Araújo JMG, Fernandes JV. Biology and pathogenesis of human osteosarcoma. Oncol Lett 2019; 19:1099-1116. [PMID: 31966039 PMCID: PMC6955653 DOI: 10.3892/ol.2019.11229] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/10/2019] [Indexed: 12/26/2022] Open
Abstract
Osteosarcoma (OS) is a bone tumor of mesenchymal origin, most frequently occurring during the rapid growth phase of long bones, and usually located in the epiphyseal growth plates of the femur or the tibia. Its most common feature is genome disorganization, aneuploidy with chromosomal alterations, deregulation of tumor suppressor genes and of the cell cycle, and an absence of DNA repair. This suggests the involvement of surveillance failures, DNA repair or apoptosis control during osteogenesis, allowing the survival of cells which have undergone alterations during differentiation. Epigenetic events, including DNA methylation, histone modifications, nucleosome remodeling and expression of non-coding RNAs have been identified as possible risk factors for the tumor. It has been reported that p53 target genes or those genes that have their activity modulated by p53, in addition to other tumor suppressor genes, are silenced in OS-derived cell lines by hypermethylation of their promoters. In osteogenesis, osteoblasts are formed from pluripotent mesenchymal cells, with potential for self-renewal, proliferation and differentiation into various cell types. This involves complex signaling pathways and multiple factors. Any disturbance in this process can cause deregulation of the differentiation and proliferation of these cells, leading to the malignant phenotype. Therefore, the origin of OS seems to be multifactorial, involving the deregulation of differentiation of mesenchymal cells and tumor suppressor genes, activation of oncogenes, epigenetic events and the production of cytokines.
Collapse
Affiliation(s)
| | | | | | | | | | - Christiane Medeiros Bezerra
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, 59072-970 Natal, RN, Brazil
| | - Vânia Sousa Andrade
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, 59072-970 Natal, RN, Brazil
| | | | - José Veríssimo Fernandes
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, 59072-970 Natal, RN, Brazil
| |
Collapse
|
17
|
Viera GM, Salomao KB, de Sousa GR, Baroni M, Delsin LEA, Pezuk JA, Brassesco MS. miRNA signatures in childhood sarcomas and their clinical implications. Clin Transl Oncol 2019; 21:1583-1623. [PMID: 30949930 DOI: 10.1007/s12094-019-02104-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
Abstract
Progresses in multimodal treatments have significantly improved the outcomes for childhood cancer. Nonetheless, for about one-third of patients with Ewing sarcoma, rhabdomyosarcoma, or osteosarcoma steady remission has remained intangible. Thus, new biomarkers to improve early diagnosis and the development of precision-targeted medicine remain imperative. Over the last decade, remarkable progress has been made in the basic understanding of miRNAs function and in interpreting the contribution of their dysregulation to cancer development and progression. On this basis, this review focuses on what has been learned about the pivotal roles of miRNAs in the regulation of key genes implicated in childhood sarcomas.
Collapse
Affiliation(s)
- G M Viera
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - K B Salomao
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - G R de Sousa
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - M Baroni
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - L E A Delsin
- Ribeirao Preto School of Medicine, University of Sao Paulo, Ribeirao Preto, Brasil
| | - J A Pezuk
- Anhanguera University of Sao Paulo, UNIAN/SP, Sao Paulo, Brasil
| | - M S Brassesco
- Faculty of Philosophy, Sciences and Letters at Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brasil.
- Departamento de Biologia, FFCLRP-USP, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirao Preto, SP, CEP 14040-901, Brazil.
| |
Collapse
|
18
|
Shreya S, Malavika D, Priya VR, Selvamurugan N. Regulation of Histone Deacetylases by MicroRNAs in Bone. Curr Protein Pept Sci 2019; 20:356-367. [PMID: 30381072 DOI: 10.2174/1389203720666181031143129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 02/08/2023]
Abstract
Formation of new bone by osteoblasts is mediated via the activation of signaling pathways, such as TGF-β, BMP, and Wnt. A number of transcription factors participate in the signaling cascades that are tightly regulated by other regulatory factors. Histone deacetylases (HDACs) are one such class of regulatory factors that play an essential role in influencing chromatin architecture and regulate the expression of the genes that play a role in osteoblast differentiation by the mechanism of deacetylation. Four classes of HDACs have been identified namely, class I, class II A, class II B, class III and class IV. MicroRNAs (miRNAs) are small fragments of non-coding RNAs typically 19-25 nucleotides long that target mRNAs to upregulate or downregulate gene expression at a post-transcriptional level. A number of miRNAs that target HDACs in bone have been recently reported. Hence, in this review, we elaborate on the various miRNAs that target the different classes of HDACs and impact of the same on osteogenesis.
Collapse
Affiliation(s)
- S Shreya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - D Malavika
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - V Raj Priya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
19
|
Saliminejad K, Khorram Khorshid HR, Soleymani Fard S, Ghaffari SH. An overview of microRNAs: Biology, functions, therapeutics, and analysis methods. J Cell Physiol 2018; 234:5451-5465. [PMID: 30471116 DOI: 10.1002/jcp.27486] [Citation(s) in RCA: 1175] [Impact Index Per Article: 167.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are a class of small noncoding RNAs, which function in posttranscriptional regulation of gene expression. They are powerful regulators of various cellular activities including cell growth, differentiation, development, and apoptosis. They have been linked to many diseases, and currently miRNA-mediated clinical trial has shown promising results for treatment of cancer and viral infection. This review provides an overview and update on miRNAs biogenesis, regulation of miRNAs expression, their biological functions, and role of miRNAs in epigenetics and cell-cell communication. In addition, alteration of miRNAs following exercise, their association with diseases, and therapeutic potential will be explained. Finally, miRNA bioinformatics tools and conventional methods for miRNA detection and quantification will be discussed.
Collapse
Affiliation(s)
- Kioomars Saliminejad
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shahrzad Soleymani Fard
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hamidollah Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Zhou L, Xu X, Liu H, Hu X, Zhang W, Ye M, Zhu X. Prognosis Analysis of Histone Deacetylases mRNA Expression in Ovarian Cancer Patients. J Cancer 2018; 9:4547-4555. [PMID: 30519361 PMCID: PMC6277648 DOI: 10.7150/jca.26780] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/09/2018] [Indexed: 12/14/2022] Open
Abstract
Histone deacetylases modulate the dynamic balance of histone acetylation and deacetylation in cells, which participate in epigenetic regulations. Accumulated evidence has demonstrated that histone deacetylases are associated with angiogenesis, cell proliferation and survival in a variety of human cancers. However, the expression and distinct prognostic value of histone deacetylases in ovarian cancer have not been well elucidated. In the present study, we collected the overall survival (OS), progress free survival (PFS), and histone deacetylases (HDAC1-11) mRNA expression in ovarian cancer from the Kaplan-Meier plotter online database. We investigated the relationship between histone deacetylases mRNA level and the clinicopathological parameters of the ovarian cancer patients, such as histology subtypes, clinical stages, grades and TP53 mutation. Our analysis data showed that over-expression of HDAC1, HDAC2, HDAC4, HDAC5 and HDAC11 were correlated to poor overall survival and unfavorable progress free survival in all ovarian cancer patients. Notably, the higher level of HDAC11 was associated with the worse OS and PFS for serous/ stage III+IV/ grade III/ TP53 mutation ovarian cancer patients. In conclusion, HDACs may play a crucial role in the prognosis of ovarian cancer, but it is worth noting that HDAC11 may be a biomarker for poor prognosis in ovarian cancer patients.
Collapse
Affiliation(s)
- Lulu Zhou
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaohui Xu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Hailing Liu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaoli Hu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Wenwen Zhang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Miaomiao Ye
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
21
|
Sun Y, Qin B. Long noncoding RNA MALAT1 regulates HDAC4-mediated proliferation and apoptosis via decoying of miR-140-5p in osteosarcoma cells. Cancer Med 2018; 7:4584-4597. [PMID: 30094957 PMCID: PMC6144160 DOI: 10.1002/cam4.1677] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 06/18/2018] [Indexed: 12/11/2022] Open
Abstract
Noncoding RNAs regulate the initiation and progression of osteosarcoma (OS). The role of long noncoding RNA metastasis‐associated lung adenocarcinoma transcript 1 (MALAT1) playing in OS and whether the function it working out was achieved through HDAC4 pathway remain uncovered. In this study, we illustrated that MALAT1 was upregulated and was correlated with poor prognosis in OS patients. Meanwhile, we demonstrated that a depression of MALAT1 suppressed proliferation and promoted apoptosis in OS cell line HOS and 143B. Further, we verified that MALAT1 exerting its function via upregulating of histone deacetylase 4 (HDAC4). Through an online prediction, a series of luciferase assays and RNA pull‐down assays, we demonstrated that both MALAT1 and HDAC4 were the targets of microRNA‐140‐5p (miR‐140‐5p) via sharing a similar microRNA responding elements. Even further, we revealed that MALAT1 served as a ceRNA of HDAC4 via decoying of miR‐140‐5p. Finally, we proved that MALAT1 promoted OS tumor growth in an in vivo animal study. In summary, the outcomes of this study demonstrated the complex ceRNA network among MALAT, miR‐140‐5p, and HDAC4‐mediated proliferation and apoptosis in OS. This study might provide a new axial in molecular treatment of OS.
Collapse
Affiliation(s)
- Yuxiu Sun
- Department of Internal Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Baoli Qin
- Department of Internal Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
22
|
Wu G, Yu W, Zhang M, Yin R, Wu Y, Liu Q. MicroRNA-145-3p suppresses proliferation and promotes apotosis and autophagy of osteosarcoma cell by targeting HDAC4. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:579-586. [PMID: 29893594 DOI: 10.1080/21691401.2018.1464459] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Studies have shown that miR-145-3p functions as a tumor suppressor and is associated with tumor growth and metastasis. This study intends to uncover the mechanism of a tumor suppressor of miR-145-3p. The expressions of miR-194 in osteosarcoma cell lines and tissues were monitored by real-time PCR. The proliferation ability was examined by MTT assay. The apoptosis and autophagy of cells were monitored by flow cytometry and microcopy, respectively. The regulation of miR-145-3p on HDAC4 was determined by luciferase assays and western blot assay. The results showed that miR-145-3p was significantly reduced in the osteosarcoma compared with the normal bone tissue. Overexpression of miR-145-3p significantly attenuated the proliferation and induced the apoptosis and autophagy of osteosarcoma cells. Furthermore, we demonstrated that miR-145-3p has inhibited the malignant behavior of osteosarcoma by down-regulating HDAC4 expression. These findings suggested that miR-145-3p may act as a tumor suppressor in osteosarcoma. MiR-145-3p/HDAC4 may be a novel therapeutic target in treatment of osteosarcoma.
Collapse
Affiliation(s)
- Guangzhi Wu
- a Department of Hand Surgery , China-Japan Union Hospital, Jilin University , Changchun , China
| | - Wei Yu
- a Department of Hand Surgery , China-Japan Union Hospital, Jilin University , Changchun , China
| | - Minglei Zhang
- b Department of Orthopedics , China-Japan Union Hospital, Jilin University , Changchun , China
| | - Ruofeng Yin
- b Department of Orthopedics , China-Japan Union Hospital, Jilin University , Changchun , China
| | - Yan Wu
- c Department of Molecular Biology , Medical College, Suchow University , Suzhou , China
| | - Qiang Liu
- c Department of Molecular Biology , Medical College, Suchow University , Suzhou , China
| |
Collapse
|
23
|
Ji X, Wang E, Tian F. MicroRNA-140 suppresses osteosarcoma tumor growth by enhancing anti-tumor immune response and blocking mTOR signaling. Biochem Biophys Res Commun 2018; 495:1342-1348. [DOI: 10.1016/j.bbrc.2017.11.120] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/19/2017] [Indexed: 12/19/2022]
|
24
|
Li H, Guan SB, Lu Y, Wang F. MiR-140-5p inhibits synovial fibroblasts proliferation and inflammatory cytokines secretion through targeting TLR4. Biomed Pharmacother 2017; 96:208-214. [DOI: 10.1016/j.biopha.2017.09.079] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/11/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022] Open
|
25
|
MiR-140/BDNF axis regulates normal human astrocyte proliferation and LPS-induced IL-6 and TNF-α secretion. Biomed Pharmacother 2017; 91:899-905. [DOI: 10.1016/j.biopha.2017.05.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 01/12/2023] Open
|