1
|
Wen R, Huang R, Xu K, Yi X. Insights into the role of histone lysine demethylases in bone homeostasis and skeletal diseases: A review. Int J Biol Macromol 2025; 306:141807. [PMID: 40054804 DOI: 10.1016/j.ijbiomac.2025.141807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 05/11/2025]
Abstract
Histone lysine demethylases (KDMs), as important epigenetic regulators, are involved in various biological processes such as energy metabolism, apoptosis, and autophagy. Recent research shows that KDMs activate or silence downstream target genes by removing lysine residues from histone tails, and participate in the regulation of bone marrow mesenchymal stem cells (BM-MSCs), osteoblasts (OB), osteoclasts (OC), chondrocytes and other skeletal cell development, differentiation and formation. Moreover, several members of the KDM family affect the occurrence and development of bone diseases such as osteoporosis (OP), osteoarthritis (OA), osteosarcoma (OS), by regulating target genes. Specific regulation mechanisms of KDMs suggest new strategies for bone disease treatment and prevention. Despite the unique function and importance of KDMs in the skeletal system, previous studies have never systematically summarized their specific role, molecular mechanism, and clinical treatment in bone physiology and pathology. Therefore, this review summarises the expression pattern, intracellular signal transduction, and mechanism of action of the KDM family in several bone physiological and pathological conditions, aiming to highlight the important role of KDMs in bone diseases and provide a reference for the future treatment of bone diseases.
Collapse
Affiliation(s)
- Ruiming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Ruiqi Huang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China; School of Physical Education, Liaoning Normal University, Dalian, Liaoning, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China.
| |
Collapse
|
2
|
Yan S, Lu T, Yang H, Ma L, Zhang Y, Li D. Decreased histone H3K9 dimethylation in synergy with DNA demethylation of Spi-1 binding site contributes to ADAMTS-5 expression in articular cartilage of osteoarthritis mice. J Cell Physiol 2024; 239:e31444. [PMID: 39318150 DOI: 10.1002/jcp.31444] [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: 06/05/2024] [Revised: 08/13/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024]
Abstract
Osteoarthritis (OA) is defined by articular cartilage degeneration, synovial membrane inflammation, and abnormal bone remodeling. Recent study has discovered that OA development is linked to an aberrant epigenetic modification of OA-related genes. Our previous research showed that DNA demethylation in ADAMTS-5 promoter region had a substantial impact on ADAMTS-5 expression in the mouse OA model. This process facilitated the binding of Spi-1 to ADAMTS-5 promoter. While alterations in histone methylation have been documented during embryonic development and cancer development, there is a paucity of data on the change in OA pathogenesis. Even no data have been reported on the role of histone modifications in ADAMTS-5 activation in OA. Following our previous study on the role of DNA methylation, we aimed to examine the contribution of histone H3K9 dimethylation in ADAMTS-5 activation in OA. Additionally, we aimed to elucidate the molecular mechanisms underlying the cooperative interaction between DNA methylation and histone H3K9 dimethylation. The potential for anti-OA intervention therapy which is based on modulating histone H3K9 dimethylation is also explored. We demonstrated that a reduction in histone H3K9 dimethylation, along with DNA demethylation of the Spi-1 binding site, had a role in ADAMTS-5 activation in the articular cartilage of OA mice. Significantly, the conditional deletion of histone demethylase to be identified as lysine-specific demethylase 1 (LSD1) in articular cartilage could alleviate the degenerative features of OA mice. Our study demonstrates the direct impact of histone H3K9 dimethylation on gene expression, which in turn contributes to OA development. This research enhances our understanding of the underlying causes of OA.
Collapse
Affiliation(s)
- Shuaichen Yan
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| | - Tongxin Lu
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| | - Huapu Yang
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| | - Liang Ma
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| | - Yuankai Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| | - Deqiang Li
- Department of Orthopedics, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, 250012, China
| |
Collapse
|
3
|
Alsabri SG, Guedi GG, Najar M, Merimi M, Lavoie F, Grabs D, Fernandes J, Pelletier JP, Martel-Pelletier J, Benderdour M, Fahmi H. Epigenetic regulation of 15-lipoxygenase-1 expression in human chondrocytes by promoter methylation. Inflamm Res 2023; 72:2145-2153. [PMID: 37874359 DOI: 10.1007/s00011-023-01805-0] [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: 08/05/2023] [Revised: 09/23/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023] Open
Abstract
OBJECTIVE AND DESIGN 15-Lipoxygenase-1 (15-LOX-1) catalyzes the biosynthesis of many anti-inflammatory and immunomodulatory lipid mediators and was reported to have protective properties in several inflammatory conditions, including osteoarthritis (OA). This study was designed to evaluate the expression of 15-LOX-1 in cartilage from normal donors and patients with OA, and to determine whether it is regulated by DNA methylation. METHODS Cartilage samples were obtained at autopsy from normal knee joints and from OA-affected joints at the time of total knee joint replacement surgery. The expression of 15-LOX-1 was evaluated using real-time polymerase chain reaction (PCR). The role of DNA methylation in 15-LOX-1 expression was assessed using the DNA methyltransferase inhibitor 5-Aza-2'-desoxycytidine (5-Aza-dC). The effect of CpG methylation on 15-LOX-1 promoter activity was evaluated using a CpG-free luciferase vector. The DNA methylation status of the 15-LOX-1 promoter was determined by pyrosequencing. RESULTS Expression of 15-LOX-1 was upregulated in OA compared to normal cartilage. Treatment with 5-Aza-dC increased 15-LOX-1 mRNA levels in chondrocytes, and in vitro methylation decreased 15-LOX-1 promoter activity. There was no difference in the methylation status of the 15-LOX-1 gene promoter between normal and OA cartilage. CONCLUSION The expression level of 15-LOX-1 was elevated in OA cartilage, which may be part of a repair process. The upregulation of 15-LOX-1 in OA cartilage was not associated with the methylation status of its promoter, suggesting that other mechanisms are involved in its upregulation.
Collapse
Affiliation(s)
- Sami G Alsabri
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Gadid G Guedi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Makram Merimi
- LBEES, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohamed Premier, Oujda, Morocco
| | - Frédéric Lavoie
- Departement of Orthopedic Surgery, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Detlev Grabs
- Department of Anatomy, Research Unit in Clinical and Functional Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Julio Fernandes
- Departement of Orthopedic Surgery, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec, H4J 1C5, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Mohamed Benderdour
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec, H4J 1C5, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada.
| |
Collapse
|
4
|
Kiełbowski K, Herian M, Bakinowska E, Banach B, Sroczyński T, Pawlik A. The Role of Genetics and Epigenetic Regulation in the Pathogenesis of Osteoarthritis. Int J Mol Sci 2023; 24:11655. [PMID: 37511413 PMCID: PMC10381003 DOI: 10.3390/ijms241411655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Osteoarthritis (OA) is progressive disease characterised by cartilage degradation, subchondral bone remodelling and inflammation of the synovium. The disease is associated with obesity, mechanical load and age. However, multiple pro-inflammatory immune mediators regulate the expression of metalloproteinases, which take part in cartilage degradation. Furthermore, genetic factors also contribute to OA susceptibility. Recent studies have highlighted that epigenetic mechanisms may regulate the expression of OA-associated genes. This review aims to present the mechanisms of OA pathogenesis and summarise current evidence regarding the role of genetics and epigenetics in this process.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (M.H.); (E.B.); (B.B.); (T.S.)
| |
Collapse
|
5
|
Assi R, Cherifi C, Cornelis FMF, Zhou Q, Storms L, Pazmino S, Coutinho de Almeida R, Meulenbelt I, Lories RJ, Monteagudo S. Inhibition of KDM7A/B histone demethylases restores H3K79 methylation and protects against osteoarthritis. Ann Rheum Dis 2023:ard-2022-223789. [PMID: 36927643 DOI: 10.1136/ard-2022-223789] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVES In osteoarthritis, methylation of lysine 79 on histone H3 (H3K79me), a protective epigenetic mechanism, is reduced. Histone methylation levels are dynamically regulated by histone methyltransferases and demethylases. Here, we aimed to identify which histone demethylases regulate H3K79me in cartilage and investigate whether their targeting protects against osteoarthritis. METHODS We determined histone demethylase expression in human non-osteoarthritis and osteoarthritis cartilage using qPCR. The role of histone demethylase families and subfamilies on H3K79me was interrogated by treatment of human C28/I2 chondrocytes with pharmacological inhibitors, followed by western blot and immunofluorescence. We performed C28/I2 micromasses to evaluate effects on glycosaminoglycans by Alcian blue staining. Changes in H3K79me after destabilisation of the medial meniscus (DMM) in mice were determined by immunohistochemistry. Daminozide, a KDM2/7 subfamily inhibitor, was intra-articularly injected in mice upon DMM. Histone demethylases targeted by daminozide were individually silenced in chondrocytes to dissect their role on H3K79me and osteoarthritis. RESULTS We documented the expression signature of histone demethylases in human non-osteoarthritis and osteoarthritis articular cartilage. Inhibition of Jumonji-C demethylase family increased H3K79me in human chondrocytes. Blockade of KDM2/7 histone demethylases with daminozide increased H3K79me and glycosaminoglycans. In mouse articular cartilage, H3K79me decayed rapidly upon induction of joint injury. Early and sustained intra-articular treatment with daminozide enhanced H3K79me and exerted protective effects in mice upon DMM. Individual silencing of KDM7A/B demethylases in human chondrocytes demonstrated that KDM7A/B mediate protective effects of daminozide on H3K79me and osteoarthritis. CONCLUSION Targeting KDM7A/B histone demethylases could be an attractive strategy to protect joints against osteoarthritis.
Collapse
Affiliation(s)
- Reem Assi
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Chahrazad Cherifi
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium .,Glycobiology Cell Growth Tissue Repair and Regeneration Research Unit, Gly-CRRET, Univ Paris Est Créteil, Créteil, France
| | - Frederique M F Cornelis
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Qiongfei Zhou
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Lies Storms
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Sofia Pazmino
- Development and Regeneration, Skeletal Biology and Engineering Research Centre, KU Leuven, Leuven, Belgium
| | - Rodrigo Coutinho de Almeida
- Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid Meulenbelt
- Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.,Integrated research on Developmental determinants of Ageing and Longevity (IDEAL), Leiden University Medical Center, Leiden, The Netherlands
| | - Rik J Lories
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Silvia Monteagudo
- Development and Regeneration, Skeletal Biology and Engineering Research Center, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Lafont JE, Moustaghfir S, Durand AL, Mallein-Gerin F. The epigenetic players and the chromatin marks involved in the articular cartilage during osteoarthritis. Front Physiol 2023; 14:1070241. [PMID: 36733912 PMCID: PMC9887161 DOI: 10.3389/fphys.2023.1070241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
Epigenetics defines the modifications of the genome that do not involve a change in the nucleotide sequence of DNA. These modifications constitute a mechanism of gene regulation poorly explored in the context of cartilage physiology. They are now intensively studied by the scientific community working on articular cartilage and its related pathology such as osteoarthritis. Indeed, epigenetic regulations can control the expression of crucial gene in the chondrocytes, the only resident cells of cartilage. Some epigenetic changes are considered as a possible cause of the abnormal gene expression and the subsequent alteration of the chondrocyte phenotype (hypertrophy, proliferation, senescence…) as observed in osteoarthritic cartilage. Osteoarthritis is a joint pathology, which results in impaired extracellular matrix homeostasis and leads ultimately to the progressive destruction of cartilage. To date, there is no pharmacological treatment and the exact causes have yet to be defined. Given that the epigenetic modifying enzymes can be controlled by pharmacological inhibitors, it is thus crucial to describe the epigenetic marks that enable the normal expression of extracellular matrix encoding genes, and those associated with the abnormal gene expression such as degradative enzyme or inflammatory cytokines encoding genes. In this review, only the DNA methylation and histone modifications will be detailed with regard to normal and osteoarthritic cartilage. Although frequently referred as epigenetic mechanisms, the regulatory mechanisms involving microRNAs will not be discussed. Altogether, this review will show how this nascent field influences our understanding of the pathogenesis of OA in terms of diagnosis and how controlling the epigenetic marks can help defining epigenetic therapies.
Collapse
|
7
|
Wang W, Ou Z, Peng J, Wang N, Zhou Y. Bioinformatics-based analysis of potential candidates chromatin regulators for immune infiltration in osteoarthritis. BMC Musculoskelet Disord 2022; 23:1123. [PMID: 36550476 PMCID: PMC9783407 DOI: 10.1186/s12891-022-06098-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Through the bioinformatics analysis to screen out the potential chromatin regulators (CRs) under the immune infiltration of osteoarthritis (OA), thus providing some theoretical support for future studies of epigenetic mechanisms under OA immune infiltration. METHODS By integrating CRs and the OA gene expression matrix, we performed weighted gene co-expression network analysis (WGCNA), differential analysis, and further screened Hub genes by protein-protein interaction (PPI) analysis. Using the OA gene expression matrix, immune infiltration extraction and quantification were performed to analyze the correlations and differences between immune infiltrating cells and their functions. By virtue of these Hub genes, Hub gene association analysis was completed and their upstream miRNAs were predicted by the FunRich software. Moreover, a risk model was established to analyze the risk probability of associated CRs in OA, and the confidence of the results was validated by the validation dataset. RESULTS This research acquired a total of 32 overlapping genes, and 10 Hub genes were further identified. The strongest positive correlation between dendritic cells and mast cells and the strongest negative correlation between parainflammation and Type I IFN reponse. In the OA group DCs, iDCs, macrophages, MCs, APC co-inhibition, and CCR were significantly increased, whereas B cells, NK cells, Th2 cells, TIL, and T cell co-stimulation were significantly decreased. The risk model results revealed that BRD1 might be an independent risk factor for OA, and the validation dataset results are consistent with it. 60 upstream miRNAs of OA-related CRs were predicted by the FunRich software. CONCLUSION This study identified certain potential CRs and miRNAs that could regulate OA immunity, thus providing certain theoretical supports for future epigenetic mechanism studies on the immune infiltration of OA.
Collapse
Affiliation(s)
- Weiwei Wang
- Guilin Hospital of Traditional Chinese Medicine, Guilin, 541002 Guangxi China
| | - Zhixue Ou
- Guilin Hospital of Traditional Chinese Medicine, Guilin, 541002 Guangxi China
| | - Jianlan Peng
- grid.256609.e0000 0001 2254 5798Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530001 Guangxi China
| | - Ning Wang
- grid.511973.8The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, 530001 Guangxi China
| | - Yi Zhou
- grid.256609.e0000 0001 2254 5798Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530001 Guangxi China
| |
Collapse
|
8
|
Tong L, Yu H, Huang X, Shen J, Xiao G, Chen L, Wang H, Xing L, Chen D. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res 2022; 10:60. [PMID: 36127328 PMCID: PMC9489702 DOI: 10.1038/s41413-022-00226-9] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/27/2022] [Accepted: 06/19/2022] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease that causes painful swelling and permanent damage to the joints in the body. The molecular mechanisms of OA are currently unknown. OA is a heterogeneous disease that affects the entire joint, and multiple tissues are altered during OA development. To better understand the pathological mechanisms of OA, new approaches, methods, and techniques need to be used to understand OA pathogenesis. In this review, we first focus on the epigenetic regulation of OA, with a particular focus on DNA methylation, histone modification, and microRNA regulation, followed by a summary of several key mediators in OA-associated pain. We then introduce several innovative techniques that have been and will continue to be used in the fields of OA and OA-associated pain, such as CRISPR, scRNA sequencing, and lineage tracing. Next, we discuss the timely updates concerning cell death regulation in OA pathology, including pyroptosis, ferroptosis, and autophagy, as well as their individual roles in OA and potential molecular targets in treating OA. Finally, our review highlights new directions on the role of the synovial lymphatic system in OA. An improved understanding of OA pathogenesis will aid in the development of more specific and effective therapeutic interventions for OA.
Collapse
Affiliation(s)
- Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China
| | - Huan Yu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xingyun Huang
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jie Shen
- Department of Orthopedic Surgery, School of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huaiyu Wang
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lianping Xing
- Department of Pathology and Laboratory of Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518005, China.
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Li J, Yang X, Chu Q, Xie L, Ding Y, Xu X, Timko MP, Fan L. Multi-omics molecular biomarkers and database of osteoarthritis. Database (Oxford) 2022; 2022:6631109. [PMID: 35788653 PMCID: PMC9254640 DOI: 10.1093/database/baac052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/15/2022] [Accepted: 06/28/2022] [Indexed: 12/05/2022]
Abstract
Osteoarthritis (OA) is the most common form of arthritis in the adult population and is a leading cause of disability. OA-related genetic loci may play an important role in clinical diagnosis and disease progression. With the rapid development of diverse technologies and omics methods, many OA-related public data sets have been accumulated. Here, we retrieved a diverse set of omics experimental results from 159 publications, including genome-wide association study, differentially expressed genes and differential methylation regions, and 2405 classified OA-related gene markers. Meanwhile, based on recent single-cell RNA-seq data from different joints, 5459 cell-type gene markers of joints were collected. The information has been integrated into an online database named OAomics and molecular biomarkers (OAOB). The database (http://ibi.zju.edu.cn/oaobdb/) provides a web server for OA marker genes, omics features and so on. To our knowledge, this is the first database of molecular biomarkers for OA.
Collapse
Affiliation(s)
- Jianhua Li
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Xiaotian Yang
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Qinjie Chu
- Institute of Bioinformatics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lingjuan Xie
- Institute of Bioinformatics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yuwen Ding
- Institute of Bioinformatics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaoxu Xu
- Institute of Bioinformatics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Michael P Timko
- Department of Biology, University of Virginia, and Department of Public Health Sciences, UVA School of Medicine, Charlottesville, VA 22904, USA
| | - Longjiang Fan
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China.,Institute of Bioinformatics, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
10
|
Luo JF, Yao YD, Cheng CS, Lio CK, Liu JX, Huang YF, He F, Xie Y, Liu L, Liu ZQ, Zhou H. Sinomenine increases the methylation level at specific GCG site in mPGES-1 promoter to facilitate its specific inhibitory effect on mPGES-1. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194813. [PMID: 35417776 DOI: 10.1016/j.bbagrm.2022.194813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/08/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
Prostaglandin E2 (PGE2) in cancer and inflammatory diseases is a key mediator of disease progression. Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used to inhibit the expression of PGE2 by depressing cyclooxygenase (COX) in inflammatory treatments. However, the inhibition to COXs may cause serious side effects. Thus, it is urgent to develop new anti-inflammatory drugs aiming new targets to inhibit PGE2 production. Microsomal prostaglandin E synthase 1 (mPGES-1) catalyzes the final step of PGE2 biosynthesis. Therefore, the selective inhibition of mPGES-1 has become a promising strategy in the treatments of cancer and inflammatory diseases. Our previous studies confirmed that sinomenine (SIN) is a specific mPGES-1 inhibitor. However, the exact mechanism by which SIN inhibits mPGES-1 remains unknown. This study aimed to explain the regulation effect of SIN to mPGES-1 gene expression by its DNA methylation induction effect. We found that the demethylating agent 5-azacytidine (5-AzaC) reversed the inhibitory effect of SIN to mPGES-1. Besides, SIN selectively increased the methylation level of the promoter region in the mPGES-1 gene while the pretreatment of 5-AzaC suppressed this effect. The results also shows that pretreatment with SIN increased the methylation level of specific GCG sites in the promoter region of mPGES-1. This specific methylation site may become a new biomarker for predicting and diagnosing RA and cancer with high expression of mPGES-1. Also, our research provides new ideas and solutions for clinical diagnosis and treatment of diseases related to mPGES-1 and for targeted methylation strategy in drug development.
Collapse
Affiliation(s)
- Jin-Fang Luo
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guian District, Guiyang, Guizhou, PR China
| | - Yun-Da Yao
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China
| | - Chun-Song Cheng
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Key Laboratory of Plant Ex-situ Conservation and Research Center of Resource Plant, Lushan Botanical Garden, Chinese Academy of Science, Jiujiang City, Jiangxi Province, PR China
| | - Chon-Kit Lio
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, Hunan, PR China
| | - Yu-Feng Huang
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, Guangdong, PR China
| | - Fan He
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, Guangdong, PR China
| | - Ying Xie
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, Guangdong, PR China.
| | - Liang Liu
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, Guangdong, PR China.
| | - Zhong-Qiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China.
| | - Hua Zhou
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macao, PR China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, Guangdong, PR China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
11
|
Wan C, Zhang F, Yao H, Li H, Tuan RS. Histone Modifications and Chondrocyte Fate: Regulation and Therapeutic Implications. Front Cell Dev Biol 2021; 9:626708. [PMID: 33937229 PMCID: PMC8085601 DOI: 10.3389/fcell.2021.626708] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
The involvement of histone modifications in cartilage development, pathology and regeneration is becoming increasingly evident. Understanding the molecular mechanisms and consequences of histone modification enzymes in cartilage development, homeostasis and pathology provides fundamental and precise perspectives to interpret the biological behavior of chondrocytes during skeletal development and the pathogenesis of various cartilage related diseases. Candidate molecules or drugs that target histone modifying proteins have shown promising therapeutic potential in the treatment of cartilage lesions associated with joint degeneration and other chondropathies. In this review, we summarized the advances in the understanding of histone modifications in the regulation of chondrocyte fate, cartilage development and pathology, particularly the molecular writers, erasers and readers involved. In addition, we have highlighted recent studies on the use of small molecules and drugs to manipulate histone signals to regulate chondrocyte functions or treat cartilage lesions, in particular osteoarthritis (OA), and discussed their potential therapeutic benefits and limitations in preventing articular cartilage degeneration or promoting its repair or regeneration.
Collapse
Affiliation(s)
- Chao Wan
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,MOE Key Laboratory for Regenerative Medicine (Shenzhen Base), School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fengjie Zhang
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,MOE Key Laboratory for Regenerative Medicine (Shenzhen Base), School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Hanyu Yao
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,MOE Key Laboratory for Regenerative Medicine (Shenzhen Base), School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Haitao Li
- MOE Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua-Peking Joint Center for Life Sciences, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Rocky S Tuan
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,MOE Key Laboratory for Regenerative Medicine (Shenzhen Base), School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
12
|
Geib T, Iacob C, Jribi R, Fernandes J, Benderdour M, Sleno L. Identification of 4-hydroxynonenal-modified proteins in human osteoarthritic chondrocytes. J Proteomics 2020; 232:104024. [PMID: 33122130 DOI: 10.1016/j.jprot.2020.104024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 10/06/2020] [Accepted: 10/22/2020] [Indexed: 11/19/2022]
Abstract
The α,β-unsaturated aldehyde 4-hydroxynonenal (HNE) is formed through lipid peroxidation during oxidative stress. As a highly reactive electrophile, it is able to form adducts with various biomolecules, including proteins. These protein modifications could modulate many signaling pathways, as well as cell differentiation and proliferation, and thus could be highly important in the context of the extracellular matrix and degradation of articular cartilage. This study specifically investigated the role of HNE as a bioactive molecule in chondrocytes of osteoarthritis (OA) patients. Chondrocyte extracts of OA and non-OA patients were analyzed for HNE binding using Western blot and bottom-up LC-MS/MS analyses. HNE-modified histones, H2A and H2B, and histone deacetylase were identified using anti-HNE antibodies. Furthermore, peptide sequencing and database searching revealed 95 distinct HNE-modified proteins and their exact modification sites, with 88 protein adducts being unique to OA chondrocytes. HNE-proteins of specific interest included histone H2A, H2B and H4, collagen alpha-3(VI) chain, eukaryotic initiation factor 4A-I, and nucleolar RNA helicase 2. Comparing their MS/MS spectra to those of HNE-modified standard peptides further validated the six HNE-proteins. SIGNIFICANCE: HNE binding to proteins has been shown to result in multiple abnormalities of chondrocyte phenotype and function, suggesting its contribution in OA development. Considering the increased levels of HNE in OA cartilage, this reactive aldehyde could play a role in OA. This work represents a clinically-relevant in vivo study to demonstrate the pathophysiological role of HNE in human OA. Since HNE binding can alter protein conformation and function, it remains highly relevant to study the effects of this modification in OA.
Collapse
Affiliation(s)
- Timon Geib
- Chemistry Department, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Cristiana Iacob
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada
| | - Rihab Jribi
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada
| | - Julio Fernandes
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada
| | - Mohamed Benderdour
- Orthopaedic Research Laboratory, Hôpital du Sacré-Coeur, University of Montréal, Montréal, Québec, Canada.
| | - Lekha Sleno
- Chemistry Department, Université du Québec à Montréal, Montréal, Québec, Canada.
| |
Collapse
|
13
|
Durand AL, Dufour A, Aubert-Foucher E, Oger-Desfeux C, Pasdeloup M, Lustig S, Servien E, Vaz G, Perrier-Groult E, Mallein-Gerin F, Lafont JE. The Lysine Specific Demethylase-1 Negatively Regulates the COL9A1 Gene in Human Articular Chondrocytes. Int J Mol Sci 2020; 21:ijms21176322. [PMID: 32878268 PMCID: PMC7504057 DOI: 10.3390/ijms21176322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of the joints which is associated with an impaired production of the cartilage matrix by the chondrocytes. Here, we investigated the role of Lysine-Specific Demethylase-1 (LSD1), a chromatin remodeling enzyme whose role in articular chondrocytes was previously associated with a catabolic activity and which is potentially involved during OA. Following a loss of function strategy and RNA sequencing analysis, we detail the genes which are targeted by LSD1 in human articular chondrocytes and identify COL9A1, a gene encoding the α1 chain of the cartilage-specific type IX collagen, as negatively regulated by LSD1. We show that LSD1 interacts with the transcription factor SOX9 and is recruited to the promoter of COL9A1. Interestingly, we observe that OA cartilage displays stronger LSD1 immunostaining compared with normal, and we demonstrate that the depletion of LSD1 in OA chondrocytes prevents the decrease in COL9A1 following Il-1β treatment. These results suggest LSD1 is a new regulator of the anabolic activity of articular chondrocytes potentially destabilizing the cartilage matrix, since it negatively regulates COL9A1, a gene encoding a crucial anchoring collagen molecule. This newly identified role played by LSD1 may thus participate in the alteration of the cartilage matrix during OA.
Collapse
Affiliation(s)
- Anne-Laure Durand
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
| | - Alexandre Dufour
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
| | - Elisabeth Aubert-Foucher
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
| | - Christine Oger-Desfeux
- PRABI-AMSB, Batiment Mendel, Campus de la Doua, Université Claude Bernard Lyon1, University Lyon, 69622 Villeurbanne CEDEX, France;
| | - Marielle Pasdeloup
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
| | - Sebastien Lustig
- FIFA Medical Center of Excellence Orthopaedic Surgery and Sports Medicine Department, Croix-Rousse Hospital, Hospices Civils de Lyon, 103 grande rue de la Croix-Rousse 69317 Lyon CEDEX 04, France and IFSTTAR, LBMC UMR_T9406 Univ Lyon, Claude Bernard Lyon 1 University, 69317 Lyon, France;
| | - Elvire Servien
- FIFA Medical Center of Excellence, Orthopaedic Surgery and Sports Medicine Department, Croix-Rousse Hospital, Hospices Civils de Lyon, 103 grande rue de la Croix-Rousse 69317 Lyon CEDEX 04, France; LIBM-EA 7424, Interuniversity Laboratory of Biology of Mobility, Claude Bernard Lyon 1 University, 69317 Lyon, France;
| | - Gualter Vaz
- Orthopaedic Surgery Department, CMCR les Massues, Croix rouge française, 92 rue Edmond Locard, 69005 Lyon, France;
| | - Emeline Perrier-Groult
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
| | - Frederic Mallein-Gerin
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
| | - Jerome E. Lafont
- CNRS UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard Lyon1, Univ Lyon, 69367 Lyon, France; (A.-L.D.); (A.D.); (E.A.-F.); (M.P.); (E.P.-G.); (F.M-G.)
- Correspondence:
| |
Collapse
|
14
|
He J, Cao W, Azeem I, Shao Z. Epigenetics of osteoarthritis: Histones and TGF-β1. Clin Chim Acta 2020; 510:593-598. [PMID: 32795546 DOI: 10.1016/j.cca.2020.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022]
Abstract
Osteoarthritis (OA) is the most common musculoskeletal and joint disorder. However, no disease-modifying therapy for OA is currently available, and the etiology of OA is poorly understood. Epigenetics has emerged as a new and important area of research on OA. Differing from genetics, Epigenetic factors are known to be tissue-specific and highly dynamic, being dependent on environmental stimuli and developmental stages. Therefore, human studies into OA epigenetics are sensitive to confounding and reverse causation. Here, we will review the epigenetic mechanism in OA onset and progression by focusing on the opposing action of two families of enzymes: histone methyltransferases and histone demethylases, such as DOT1L, KDM4B, KDM6A, KDM6B, EZH2, and LSD1. Moreover, the TGF-β1 signaling pathway has proven to be one of the key factors in cartilage and bone formation, and in recent research, was found to initiate and develop OA disease by TGF-β1 overexpression. Besides the introduction of enzymes and TGF-β1 signaling, some special epigenetic regulation mechanisms associated with key transcription factors (e.g. RUNX2, NFAT1, and SOX9) in OA disease are also reviewed here in detail to clarify the OA epigenetic mechanism. The overall understanding of these epigenetic mechanisms underlying the issues will accelerate the development of novel therapeutic strategies for OA.
Collapse
Affiliation(s)
- Jianwei He
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; The First Affiliated Hospital, Shihezi University, School of Medicine, Xinjiang, China
| | - Weiwei Cao
- Key Laboratory of Xinjiang Endemic and Ethnic Disease, Shihezi University, School of Medicine, Xinjiang, China
| | - Inayat Azeem
- Office for Education to International Students, School of Medicine, Shihezi University, Xinjiang, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
Ukita M, Matsushita K, Tamura M, Yamaguchi T. Histone H3K9 methylation is involved in temporomandibular joint osteoarthritis. Int J Mol Med 2019; 45:607-614. [PMID: 31894302 DOI: 10.3892/ijmm.2019.4446] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/11/2019] [Indexed: 11/05/2022] Open
Abstract
The morbidity of temporomandibular joint osteoarthritis (TMJOA) increases with age. Condylar articular cartilage degradation, which causes TMJOA, is known to be involved in articular chondrocyte metabolic imbalances in the temporomandibular joint (TMJ) and in other joints of the body. Epigenetic regulation, such as the chemical modification of DNA and histones, is implicated in cartilage homeostasis. However, few studies have been conducted on the epigenetic regulation of condylar articular cartilage degradation. The present study investigated the regulation of histone H3 lysine 9 (H3K9) methylation and its effects on the pathogenesis of degenerative TMJ cartilage disorders. The histone H3K9 methylation level was decreased in degenerated condylar articular cartilage in aged mice. Treatment with chaetocin (a selective H3K9 methylation inhibitor) reduced cell viability and promoted caspase‑3/7 activity in ATDC5 mouse chondroprogenitor cells. The inhibition of H3K9 methylation increased matrix metalloproteinase (Mmp)1 and Mmp13 mRNA expression in these cells. Furthermore, the expression levels of Sox9 and collagen α1(II) (Col2a1) mRNA, which are anabolic factors for chondrogenic differentiation, were also decreased by treatment with chaetocin, which is an inhibitor of histone methyltransferases. These results indicated that histone H3K9 methylation regulates chondrocyte homeostasis in terms of cell growth, apoptosis and gene expression, and highlighted a possible future therapy option for TMJOA.
Collapse
Affiliation(s)
- Mayumi Ukita
- Crown and Bridge Prosthodontics, Department of Oral Functional Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8586, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi 474‑8511, Japan
| | - Masato Tamura
- Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8586, Japan
| | - Taihiko Yamaguchi
- Crown and Bridge Prosthodontics, Department of Oral Functional Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060‑8586, Japan
| |
Collapse
|
16
|
Valès S, Bacola G, Biraud M, Touvron M, Bessard A, Geraldo F, Dougherty KA, Lashani S, Bossard C, Flamant M, Duchalais E, Marionneau-Lambot S, Oullier T, Oliver L, Neunlist M, Vallette FM, Van Landeghem L. Tumor cells hijack enteric glia to activate colon cancer stem cells and stimulate tumorigenesis. EBioMedicine 2019; 49:172-188. [PMID: 31662289 PMCID: PMC6945247 DOI: 10.1016/j.ebiom.2019.09.045] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Colon cancer stem cells (CSCs), considered responsible for tumor initiation and cancer relapse, are constantly exposed to regulatory cues emanating from neighboring cells present in the tumor microenvironment. Among these cells are enteric glial cells (EGCs) that are potent regulators of the epithelium functions in a healthy intestine. However, whether EGCs impact CSC-driven tumorigenesis remains unknown. METHODS Impact of human EGC primary cultures or a non-transformed EGC line on CSCs isolated from human primary colon adenocarcinomas or colon cancer cell lines with different p53, MMR system and stemness status was determined using murine xenograft models and 3D co-culture systems. Supernatants of patient-matched human primary colon adenocarcinomas and non-adjacent healthy mucosa were used to mimic tumor versus healthy mucosa secretomes and compare their effects on EGCs. FINDINGS Our data show that EGCs stimulate CSC expansion and ability to give rise to tumors via paracrine signaling. Importantly, only EGCs that were pre-activated by tumor epithelial cell-derived soluble factors increased CSC tumorigenicity. Pharmacological inhibition of PGE2 biosynthesis in EGCs or IL-1 knockdown in tumor epithelial cells prevented EGC acquisition of a pro-tumorigenic phenotype. Inhibition of PGE2 receptor EP4 and EGFR in CSCs inhibited the effects of tumor-activated EGCs. INTERPRETATION Altogether, our results show that EGCs, once activated by the tumor, acquire a pro-tumorigenic phenotype and stimulate CSC-driven tumorigenesis via a PGE2/EP4/EGFR-dependent pathway. FUNDING This work was supported by grants from the French National Cancer Institute, La Ligue contre le Cancer, the 'Région des Pays de la Loire' and the UNC Lineberger Comprehensive Cancer Center.
Collapse
Affiliation(s)
- Simon Valès
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Gregory Bacola
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Mandy Biraud
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Mélissa Touvron
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Anne Bessard
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Fanny Geraldo
- Nantes University, INSERM 1232, CRCINA, Nantes, France
| | - Kelsie A. Dougherty
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Shaian Lashani
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | | | - Mathurin Flamant
- Nantes University Hospital, Nantes, France,Jules Verne Clinic, Nantes, France
| | - Emilie Duchalais
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Nantes University Hospital, Nantes, France
| | | | | | - Lisa Oliver
- Nantes University, INSERM 1232, CRCINA, Nantes, France,Nantes University Hospital, Nantes, France
| | - Michel Neunlist
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Nantes University Hospital, Nantes, France
| | | | - Laurianne Van Landeghem
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA,Corresponding author at: North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, CB# 8401, Raleigh, NC 27607, USA.
| |
Collapse
|
17
|
Chen Z, Cai X, Li M, Yan L, Wu L, Wang X, Tang N. CRISPR/Cas9-based liver-derived reporter cells for screening of mPGES-1 inhibitors. J Enzyme Inhib Med Chem 2019; 34:799-807. [PMID: 30879343 PMCID: PMC6427568 DOI: 10.1080/14756366.2019.1587416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
mPGES-1 is a terminal rate-limiting enzyme responsible for inflammation-induced PGE2 production. The inhibition of mPGES-1 has been considered as a safe and effective target for the treatment of inflammation and cancer. However, a specific, efficient, and simple method for high-throughput screening of mPGES-1 inhibitors is still lacking. In this study, we developed a fluorescence imaging strategy to monitor the expression of mPGES-1 via CRISPR/Cas9 knock-in system. Immunofluorescence colocalisation, Sanger sequencing, RNAi, and IL-1β treatment all confirmed the successful construction of mPGES-1 reporter cells. The fluorescence signal intensity of the reporter cells treated with four conventional mPGES-1 inhibitors was considerably attenuated via flow cytometry and fluorescent microplate reader, demonstrating that the reporter cells can be used as an efficient and convenient means for screening and optimising mPGES-1 inhibitors. Moreover, it provides a new technical support for the development of targeted small molecule compounds for anti-inflammatory and tumour therapy.
Collapse
Affiliation(s)
- Zhanfei Chen
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China
| | - Xiaoling Cai
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China
| | - Man Li
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China
| | - LinLin Yan
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China
| | - Luxi Wu
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China
| | - Xiaoqian Wang
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China
| | - Nanhong Tang
- a Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital , Fuzhou , China.,b Key Laboratory of Ministry of Education for Gastrointestinal Cancer , Research Center for Molecular Medicine, Fujian Medical University , Fuzhou , China
| |
Collapse
|
18
|
Fathollahi A, Aslani S, Jamshidi A, Mahmoudi M. Epigenetics in osteoarthritis: Novel spotlight. J Cell Physiol 2019; 234:12309-12324. [DOI: 10.1002/jcp.28020] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Anwar Fathollahi
- Department of Immunology School of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
19
|
Pinheiro DML, de Oliveira AHS, Coutinho LG, Fontes FL, de Medeiros Oliveira RK, Oliveira TT, Faustino ALF, Lira da Silva V, de Melo Campos JTA, Lajus TBP, de Souza SJ, Agnez-Lima LF. Resveratrol decreases the expression of genes involved in inflammation through transcriptional regulation. Free Radic Biol Med 2019; 130:8-22. [PMID: 30366059 DOI: 10.1016/j.freeradbiomed.2018.10.432] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
Abstract
Oxidative stress generated during inflammation is associated with a wide range of pathologies. Resveratrol (RESV) displays anti-inflammatory and antioxidant activities, being a candidate for the development of adjuvant therapies for several inflammatory diseases. Despite this potential, the cellular responses induced by RESV are not well known. In this work, transcriptomic analysis was performed following lipopolysaccharide (LPS) stimulation of monocyte cultures in the presence of RESV. Induction of an inflammatory response was observed after LPS treatment and the addition of RESV led to decreases in expression of the inflammatory mediators, tumor necrosis factor-alpha (TNF-α), interleukin-8 (IL-8), and monocyte chemoattractant protein-1 (MCP-1), without cytotoxicity. RNA sequencing revealed 823 upregulated and 2098 downregulated genes (cutoff ≥2.0 or ≤-2.0) after RESV treatment. Gene ontology analysis showed that the upregulated genes were associated with metabolic processes and the cell cycle, consistent with normal cell growth and differentiation under an inflammatory stimulus. The downregulated genes were associated with inflammatory responses, gene expression, and protein modification. The prediction of master regulators using the iRegulon tool showed nuclear respiratory factor 1 (NRF1) and GA-binding protein alpha subunit (GABPA) as the main regulators of the downregulated genes. Using immunoprecipitation and protein expression assays, we observed that RESV was able to decrease protein acetylation patterns, such as acetylated apurinic/apyrimidinic endonuclease-1/reduction-oxidation factor 1 (APE1/Ref-1), and increase histone methylation. In addition, reductions in p65 (nuclear factor-kappa B (NF-κB) subunit) and lysine-specific histone demethylase-1 (LSD1) expression were observed. In conclusion, our data indicate that treatment with RESV caused significant changes in protein acetylation and methylation patterns, suggesting the induction of deacetylase and reduction of demethylase activities that mainly affect regulatory cascades mediated by NF-кB and Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling. NRF1 and GABPA seem to be the main regulators of the transcriptional profile observed after RESV treatment.
Collapse
Affiliation(s)
| | - Ana Helena Sales de Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil; Chemistry Department, New York University, New York, NY, United States
| | - Leonam Gomes Coutinho
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil; Instituto Federal de Educação Tecnológica do Rio Grande do Norte, IFRN, São Paulo do Potengi, Brazil
| | - Fabrícia Lima Fontes
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil
| | | | - Thais Teixeira Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil
| | - André Luís Fonseca Faustino
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil; Bioinformatics Multidisciplinary Environment (BioME), IMD, UFRN, Brazil
| | - Vandeclécio Lira da Silva
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil; Bioinformatics Multidisciplinary Environment (BioME), IMD, UFRN, Brazil
| | | | - Tirzah Braz Petta Lajus
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, UFRN, Natal, Brazil
| | - Sandro José de Souza
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil; Bioinformatics Multidisciplinary Environment (BioME), IMD, UFRN, Brazil
| | | |
Collapse
|
20
|
Shao G, Wan X, Lai W, Wu C, Jin J, Liu X, Wei Y, Lin Q, Zhang L, Shao Q. Inhibition of lysine-specific demethylase 1 prevents proliferation and mediates cisplatin sensitivity in ovarian cancer cells. Oncol Lett 2018; 15:9025-9032. [PMID: 29928330 PMCID: PMC6004655 DOI: 10.3892/ol.2018.8511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 01/31/2018] [Indexed: 12/11/2022] Open
Abstract
Lysine-specific demethylase 1 (LSD1) functions as a transcriptional coregulator by modulating histone methylation and has been associated with numerous high-risk cancers. Previously, our group and others identified LSD1 as an upregulated gene in ovarian cancer, and reported that the upregulation of LSD1 was associated with poor prognosis of patients with ovarian cancer. However, the role of LSD1 in ovarian cancer requires further investigation. The present study revealed that the overexpression of LSD1 significantly promoted the proliferation of SKOV3 ovarian cancer cells, while knockdown of LSD1 markedly inhibited cell proliferation and potentiated cisplatin-induced cell apoptosis, supporting LSD1 as an oncogenic protein in ovarian cancer. Mechanistic studies have indicated that LSD1 modulates the expression of cyclin dependent kinase inhibitor 1, Survivin, B-cell lymphoma-2 (Bcl-2) and Bcl-2-associated X genes, which are known regulators of cell proliferation. Furthermore, LSD1 knockdown plus cisplatin synergistically impaired cell migration via the induction of the epithelial marker E-cadherin and inhibition of the mesenchymal markers, snail family transcriptional repressor 1 and Vimentin. These data of the present study indicated LSD1 as a potential regulator of ovarian cancer cell progression and suggested an unfavorable role of LSD1 in cisplatin-based regimens.
Collapse
Affiliation(s)
- Genbao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaolei Wan
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Oncology, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China.,Department of Oncology, The Affiliated Jurong Hospital, Jiangsu University, Zhenjiang, Jiangsu 212400, P.R. China
| | - Wensheng Lai
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Oncology, The Affiliated Jurong Hospital, Jiangsu University, Zhenjiang, Jiangsu 212400, P.R. China
| | - Chaoyang Wu
- Department of Oncology, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Jie Jin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiuwen Liu
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ye Wei
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qiong Lin
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Liuping Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qixiang Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
21
|
Miranda-Duarte A. DNA Methylation in Osteoarthritis: Current Status and Therapeutic Implications. Open Rheumatol J 2018; 12:37-49. [PMID: 29682093 PMCID: PMC5885469 DOI: 10.2174/1874312901812010037] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/24/2018] [Accepted: 03/05/2018] [Indexed: 01/25/2023] Open
Abstract
Background: Primary Osteoarthritis (OA) is a multifactorial disease in which genetic factors are strongly associated with its development; however, recently it has been observed that epigenetic modifications are also involved in the pathogenesis of OA. DNA methylation is related to gene silencing, and several studies have investigated its role in the loci of different pathways or molecules associated to OA. Objective: This review is focused on the current status of DNA methylation studies related to OA pathogenesis. Method: A review of the literature was conducted on searching in PUBMED for original papers on DNA methylation in OA. Conclusion: The DNA methylation research of loci related to OA pathogenesis has shown a correlation between methylation and gene repression; however, there are some exceptions to this rule. Recently, the development of genome-wide methylation and genome-wide hydroxymethylation profiles has demonstrated that several genes previously associated with OA can have changes in their methylation status, favoring the development of the disease, and these have even shown the role of other epigenetic markers.
Collapse
Affiliation(s)
- Antonio Miranda-Duarte
- Department of Genetics, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Tlalpan, Mexico
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Epigenomics has emerged as a key player in our rapidly evolving understanding of osteoarthritis. Historical studies implicated epigenetic alterations, particularly DNA methylation, in OA pathogenesis; however, recent technological advances have resulted in numerous epigenome-wide studies examining in detail epigenetic modifications in OA. The purpose of this article is to introduce basic concepts in epigenetics and their recent applications to the study of osteoarthritis development and progression. RECENT FINDINGS Epigenetics describes three major phenomena: DNA modification via methylation, histone sidechain modifications, and short noncoding RNA sequences which work in concert to regulate gene transcription in a heritable fashion. Cartilage has been the most widely studied tissue in OA, and differential methylation of genes involved in inflammation, cell cycle, TGFβ, and HOX genes have been confirmed several times. Bone studies suggest similar findings, and the intriguing possibility of epigenetic changes in subchondral bone during many OA processes. Multiple studies have demonstrated the involvement of certain noncoding RNAs, particularly miR-140, in OA development via modulation of key catabolic factors. Although much work has been done, much is still unknown. Future epigenomic studies will no doubt continue to widen our understanding of extraarticular tissues and OA pathogenesis, and studies in animal models may offer glimpses into epigenome alterations in the earliest stages of OA.
Collapse
|
23
|
Chen J, Ding J, Wang Z, Zhu J, Wang X, Du J. Identification of downstream metastasis-associated target genes regulated by LSD1 in colon cancer cells. Oncotarget 2017; 8:19609-19630. [PMID: 28121627 PMCID: PMC5386709 DOI: 10.18632/oncotarget.14778] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/12/2016] [Indexed: 12/29/2022] Open
Abstract
Purpose This study aims to identify downstream target genes regulated by lysine-specific demethylase 1 (LSD1) in colon cancer cells and investigate the molecular mechanisms of LSD1 influencing invasion and metastasis of colon cancer. Method We obtained the expression changes of downstream target genes regulated by small-interfering RNA-LSD1 and LSD1-overexpression via gene expression profiling in two human colon cancer cell lines. An Affymetrix Human Transcriptome Array 2.0 was used to identify differentially expressed genes (DEGs). We screened out LSD1-target gene associated with proliferation, metastasis, and invasion from DEGs via Gene Ontology and Pathway Studio. Subsequently, four key genes (CABYR, FOXF2, TLE4, and CDH1) were computationally predicted as metastasis-related LSD1-target genes. ChIp-PCR was applied after RT-PCR and Western blot validations to detect the occupancy of LSD1-target gene promoter-bound LSD1. Result A total of 3633 DEGs were significantly upregulated, and 4642 DEGs were downregulated in LSD1-silenced SW620 cells. A total of 4047 DEGs and 4240 DEGs were upregulated and downregulated in LSD1-overexpressed HT-29 cells, respectively. RT-PCR and Western blot validated the microarray analysis results. ChIP assay results demonstrated that LSD1 might be negative regulators for target genes CABYR and CDH1. The expression level of LSD1 is negatively correlated with mono- and dimethylation of histone H3 lysine4(H3K4) at LSD1- target gene promoter region. No significant mono-methylation and dimethylation of H3 lysine9 methylation was detected at the promoter region of CABYR and CDH1. Conclusion LSD1- depletion contributed to the upregulation of CABYR and CDH1 through enhancing the dimethylation of H3K4 at the LSD1-target genes promoter. LSD1- overexpression mediated the downregulation of CABYR and CDH1expression through decreasing the mono- and dimethylation of H3K4 at LSD1-target gene promoter in colon cancer cells. CABYR and CDH1 might be potential LSD1-target genes in colon carcinogenesis.
Collapse
Affiliation(s)
- Jiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Ding
- Department of Gastrointestinal Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Zhu
- Department of Mini-invasive Surgery, Guiyang Hospital of Guizhou Aviation Industry Group, Guiyang, China
| | - Xuejian Wang
- Department of Mini-invasive Surgery, Guiyang Hospital of Guizhou Aviation Industry Group, Guiyang, China
| | - Jiyi Du
- Department of Gastrointestinal Surgery The First People's Hospital of Guiyang, Guiyang, China
| |
Collapse
|
24
|
Zhou PH, Shi L, Qiu B. Protective effect of controlled release of cytokine response modifier A from chitosan microspheres on rat chondrocytes from interleukin-1β induced inflammation and apoptosis. Exp Ther Med 2017; 14:3170-3178. [PMID: 28912867 DOI: 10.3892/etm.2017.4884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 05/11/2017] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of cytokine response modifier A (CrmA) released from chitosan (CS) microspheres in a controlled manner on interleukin (IL)-1β-induced inflammation and apoptosis in chondrocytes. The CrmA release kinetics were characterized by an initial burst release, which was reduced to a linear release over 8 days. Furthermore, chondrocytes were isolated from 1-week-old Sprague Dawley rats. The cell culture was established by stimulation with 10 ng/ml IL-1β and subsequent incubation with CS-CrmA microspheres. Following stimulation with IL-1β, the viability of chondrocytes was decreased. However, the cell viability was attenuated by CS-CrmA microspheres as revealed by a cell counting kit-8 assay. CS-CrmA microspheres significantly inhibited IL-1β-induced inflammation in chondrocytes by attenuating increases in the gene expression levels of inducible nitric oxide synthase and cyclooxygenase-2, as well as the concentrations of nitric oxide and prostaglandin E2. CS-CrmA microspheres significantly decreased the number of apoptotic chondrocytes induced by IL-1β as indicated by a terminal deoxyribonucleotide transferase deoxyuridine triphosphate nick-end labeling assay. In addition, CS-CrmA microspheres blocked IL-1β-induced chondrocyte apoptosis by increasing B-cell lymphoma 2 (Bcl-2) and decreasing Bcl-2-associated X protein, caspase-3 and poly adenosine diphosphate-ribose polymerase expression at the mRNA and protein levels, as indicated by reverse-transcription quantitative polymerase chain reaction and western blot analysis, respectively. The results of the present study revealed that CS-CrmA microspheres, as a controlled release system of CrmA, may protect rat chondrocytes from IL-1β-induced inflammation and apoptosis via regulating inflammatory and apoptosis-associated genes.
Collapse
Affiliation(s)
- Pang-Hu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lei Shi
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bo Qiu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
25
|
Hennequart M, Pilotte L, Cane S, Hoffmann D, Stroobant V, Plaen ED, Van den Eynde B. Constitutive IDO1 Expression in Human Tumors Is Driven by Cyclooxygenase-2 and Mediates Intrinsic Immune Resistance. Cancer Immunol Res 2017; 5:695-709. [PMID: 28765120 DOI: 10.1158/2326-6066.cir-16-0400] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/02/2017] [Accepted: 06/30/2017] [Indexed: 12/16/2022]
Abstract
Tumors use various mechanisms to avoid immune destruction. Cyclooxygenase-2 (COX-2) expression may be a driver of immune suppression in melanoma, but the mechanisms involved remain elusive. Here, we show that COX-2 expression drives constitutive expression of indoleamine 2,3-dioxygenase 1 (IDO1) in human tumor cells. IDO1 is an immunosuppressive enzyme that degrades tryptophan. In a series of seven human tumor lines, constitutive IDO1 expression depends on COX-2 and prostaglandin E2 (PGE2), which, upon autocrine signaling through the EP receptor, activates IDO1 via the PKC and PI3K pathways. COX-2 expression itself depends on the MAPK pathway, which therefore indirectly controls IDO1 expression. Most of these tumors carry PI3K or MAPK oncogenic mutations, which may favor constitutive IDO1 expression. Celecoxib treatment promoted immune rejection of IDO1-expressing human tumor xenografts in immunodeficient mice reconstituted with human allogeneic lymphocytes. This effect was associated with a reduced expression of IDO1 in those ovarian SKOV3 tumors and an increased infiltration of CD3+ and CD8+ cells. Our results highlight the role of COX-2 in constitutive IDO1 expression by human tumors and substantiate the use of COX-2 inhibitors to improve the efficacy of cancer immunotherapy, by reducing constitutive IDO1 expression, which contributes to the lack of T-cell infiltration in "cold" tumors, which fail to respond to immunotherapy. Cancer Immunol Res; 5(8); 695-709. ©2017 AACR.
Collapse
Affiliation(s)
- Marc Hennequart
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Luc Pilotte
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Stefania Cane
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Delia Hoffmann
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Etienne De Plaen
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Benoît Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium. .,de Duve Institute, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| |
Collapse
|
26
|
Ma Z, Wang Y, Piao T, Liu J. Echinocystic Acid Inhibits IL-1β-Induced COX-2 and iNOS Expression in Human Osteoarthritis Chondrocytes. Inflammation 2017; 39:543-9. [PMID: 26499345 DOI: 10.1007/s10753-015-0278-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Echinocystic acid (EA), a pentacyclic triterpene isolated from the fruits of Gleditsia sinensis Lam, displays a range of pharmacological activities including anti-inflammatory and antioxidant effects. However, the effect of EA on IL-1β-stimulated osteoarthritis chondrocyte has not been reported. The purpose of this study was to assess the effects of EA on IL-1β-stimulated human osteoarthritis chondrocyte. Chondrocytes were stimulated with IL-1β in the absence or presence of EA. NO and PGE2 production were measured by Griess reagent and ELISA. The expression of COX-2, iNOS, nuclear factor-κB (NF-κB), inhibitory kappa B (IκBα), c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK) were detected by Western blot analysis. The results showed that EA suppressed IL-1β-induced collagenase-3 (MMP-13), NO, and PGE2 production in a dose-dependent manner. IL-1β up-regulated the expression of COX-2 and iNOS, and the increase was inhibited by EA. Furthermore, IL-1β-induced NF-κB and mitogen-activated protein kinase (MAPK) activation were inhibited by EA. In conclusion, EA effectively attenuated IL-1β-induced inflammatory response in osteoarthritis chondrocyte which suggesting that EA may be a potential agent in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Zhiqiang Ma
- Department of Orthopedic Surgery, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, People's Republic of China
| | - Yanlong Wang
- Department of Orthopedic Surgery, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, People's Republic of China
| | - Taikui Piao
- Children's Hospital of Harbin, Harbin, Heilongjiang Province, 150010, People's Republic of China
| | - Jianyu Liu
- Department of Orthopedic Surgery, The Second Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, People's Republic of China.
| |
Collapse
|
27
|
|
28
|
Abstract
Osteoarthritis (OA) was once defined as a non-inflammatory arthropathy, but it is now well-recognized that there is a major inflammatory component to this disease. In addition to synovial cells, articular chondrocytes and other cells of diarthrodial joints are also known to express inflammatory mediators. It has been proposed that targeting inflammation pathways could be a promising strategy to treat OA. There have been many reports of cross-talk between inflammation and epigenetic factors in cartilage. Specifically, inflammatory mediators have been shown to regulate levels of enzymes that catalyze changes in DNA methylation and histone structure, as well as alter levels of non-coding RNAs. In addition, expression levels of a number of these epigenetic factors have been shown to be altered in OA, thereby suggesting potential interplay between inflammation and epigenetics in this disease. This review provides information on inflammatory pathways in arthritis and summarizes published research on how epigenetic regulators are affected by inflammation in chondrocytes. Furthermore, we discuss data showing how altered expression of some of these epigenetic factors can induce either catabolic or anti-catabolic effects in response to inflammatory signals. A better understanding of how inflammation affects epigenetic factors in OA may provide us with novel therapeutic strategies to treat this condition.
Collapse
Affiliation(s)
- Jie Shen
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Regis J. O'Keefe
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Chen L, Wu Y, Wu Y, Wang Y, Sun L, Li F. The inhibition of EZH2 ameliorates osteoarthritis development through the Wnt/β-catenin pathway. Sci Rep 2016; 6:29176. [PMID: 27539752 PMCID: PMC4990905 DOI: 10.1038/srep29176] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/06/2016] [Indexed: 11/09/2022] Open
Abstract
The purpose of our study was to elucidate the role of the histone methyltransferase enhancer of zeste homologue 2 (EZH2) in the pathophysiology of osteoarthritis (OA) and to develop a strategy to modulate EZH2 activity for OA treatment. The expression of EZH2 in normal and OA human cartilage was compared by western blotting. The effect of EZH2 overexpression and inhibition on chondrocyte hypertrophy related gene expression was examined by real-time PCR, and histone methylation on the promoter of the Wnt inhibitor SFRP1 was analyzed using a chromatin immunoprecipitation (ChIP) PCR. Histological assessment of OA mice joint was carried out to assess the in vivo effects of EZH2 inhibitor EPZ005687. We found EZH2 level was significantly increased in the chondrocytes of OA patients compared to normal humans. Overexpression of EZH2 promoted Indian Hedgehog, MMP-13, ADAMTS-5 and COLX expression, while inhibition of EZH2 reversed this trend. Furthermore, the induction of EZH2 led to β-catenin signaling activation by increasing H3K27me3 on the promoter of SFRP1, while the inhibition of EZH2 silenced β-catenin signaling. Finally, intraarticular injection of EPZ005687 delayed OA development in mice. These results implicated EZH2 activity in OA development. Pharmacological inhibition of EZH2 may be an effective therapeutic approach for osteoarthritis.
Collapse
Affiliation(s)
- Linwei Chen
- Department of orthopedics, Second affiliated hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yaosen Wu
- Department of orthopedics, Second affiliated hospital, Wenzhou Medical university, Wenzhou, Zhejiang Province, China
| | - Yan Wu
- Department of orthopedics, Second affiliated hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ye Wang
- Department of pediatrics, Second affiliated hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Liaojun Sun
- Department of orthopedics, Second affiliated hospital, Wenzhou Medical university, Wenzhou, Zhejiang Province, China
| | - Fangcai Li
- Department of orthopedics, Second affiliated hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
30
|
Jin W, Peng J, Jiang S. The epigenetic regulation of embryonic myogenesis and adult muscle regeneration by histone methylation modification. Biochem Biophys Rep 2016; 6:209-219. [PMID: 28955879 PMCID: PMC5600456 DOI: 10.1016/j.bbrep.2016.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle formation in vertebrates is derived from the paraxial mesoderm, which develops into myogenic precursor cells and finally differentiates into mature myofibers. This myogenic program involves temporal-spatial molecular events performed by transcription regulators (such as members of the Pax, MRFs and Six families) and signaling pathways (such as Wnts, BMP and Shh signaling). Epigenetic regulation, including histone post-translational modifications is crucial for controlling gene expression through recruitment of various chromatin-modifying enzymes that alter chromatin dynamics during myogenesis. The chromatin modifying enzymes are also recruited at regions of muscle gene regulation, coordinating transcription regulators to influence gene expression. In particular, the reversible methylation status of histone N-terminal tails provides the important regulatory mechanisms in either activation or repression of muscle genes. In this report, we review the recent literatures to deduce mechanisms underlying the epigenetic regulation of gene expression with a focus on histone methylation modification during embryo myogenesis and adult muscle regeneration. Recent results from different histone methylation/demethylation modifications have increased our understanding about the highly intricate layers of epigenetic regulations involved in myogenesis and cross-talk of histone enzymes with the muscle-specific transcriptional machinery. Myogenesis is influenced by regulation of transcription factors, signal pathways and post-transcriptional modifications. Histone methylation modifications as “on/off” switches regulated myogenic lineage commitment and differentiation. The myogenic regulatory factors and histone methylation modifications established dynamic regulatory mechanism.
Collapse
Key Words
- BMP4, bone morphogenic protein 4
- ChIP, chromatin immunoprecipitation
- Epigenetic
- H3K27, methylation of histone H3 lysine 27
- H3K4, methylation of histone H3 lysine 4
- H3K9, methylation of histone H3 lysine 9
- Histone methylation/demethylation modification
- KDMs, lysine demethyltransferases
- LSD1, lysine specific demethyltransferase 1
- MEF2, myocyte enhancer factor 2
- MRFs, myogenic regulatory factors
- Muscle differentiation
- Muscle progenitor cells
- Muscle regeneration
- Myogenesis
- PRC2, polycomb repressive complex 2
- SCs, satellite cells
- Shh, sonic hedgehog
- TSS, transcription start sites
- UTX, ubiquitously transcribed tetratricopeptide repeat, X chromosome
- bHLH, basic helix-loop-helix
- p38 MAPK, p38 mitogen-activated protein kinase
Collapse
Affiliation(s)
- Wei Jin
- Key Laboratory of Pig Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, PR China
| | - Siwen Jiang
- Key Laboratory of Pig Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, PR China.,Key Projects in the Cooperative Innovation Center for Sustainable Pig Production of Wuhan, PR China
| |
Collapse
|
31
|
Zhang M, Lu Q, Miller AH, Barnthouse NC, Wang J. Dynamic epigenetic mechanisms regulate age-dependent SOX9 expression in mouse articular cartilage. Int J Biochem Cell Biol 2016; 72:125-134. [PMID: 26806292 DOI: 10.1016/j.biocel.2016.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/16/2015] [Accepted: 01/18/2016] [Indexed: 12/22/2022]
Abstract
While the developmental role of the SOX9 transcription factor in chondrocyte differentiation and cartilage formation is well documented, age-dependent SOX9 expression in articular chondrocytes (ACs) and its regulatory mechanisms remain unclear. This study aimed to explore epigenetic regulatory mechanisms for age-related changes in SOX9 expression in ACs of mice, spanning from the developmental stage to 18 months of age. Sox9 mRNA and protein were highly expressed in ACs during joint development but significantly decreased after 2 months of age. Histopathological features of osteoarthritis were not observed in examined hip and shoulder joints by 18 months of age. Epigenetic studies revealed that DNA methylation levels were increased at specific CpG islands of the Sox9 gene at 6 and 12 months; treatment of cultured ACs from 6-month-old mice with 5-azacytidine (an inhibitor of DNA methylation) elevated the level of Sox9 expression in ACs by lowering DNA methylation levels in the Sox9 promoter region. Histone 3 lysine 4 dimethylation (H3K4me2, a histone modification for transcriptional activation) in the Sox9 promoter region was decreased with age, which was associated with the age-dependent decrease in SOX9 expression in ACs. Knockdown of lysine-specific demethylase-1 up-regulated SOX9 expression in ACs of adult mice through increased recruitment of H3K4me2 in the Sox9 promoter region. Our results suggest that SOX9 expression in mouse ACs is significantly decreased after the completion of joint development. These age-dependent changes in SOX9 expression are dynamically regulated by DNA methylation and histone methylation.
Collapse
Affiliation(s)
- Mingcai Zhang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Qinghua Lu
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Andrew H Miller
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Nicholas C Barnthouse
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS 66160, United States; Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
32
|
Abstract
Osteoarthritis (OA) is a common degenerative joint disease, the pathological mechanism of which is currently unknown. Genetic alteration is one of the key contributing factors for OA pathology. Recent evidence suggests that epigenetic and microRNA regulation of critical genes may contribute to OA development. In this article, we review the epigenetic and microRNA regulations of genes related to OA development. Potential therapeutic strategies may be developed on the basis of novel findings.
Collapse
Affiliation(s)
- Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jie Shen
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, 63110, USA
| | - Tianqian Hui
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, 60612, USA
| |
Collapse
|