1
|
Xiang Y, Hu X, Yang X, Wang G, Li Y, Sun F, Song E. Andrographolide suppresses fibrogenic phenotype of chondrocytes and ameliorates osteoarthritis by regulating miR-137/BMP7 axis. J Orthop 2025; 64:108-116. [PMID: 39691644 PMCID: PMC11648649 DOI: 10.1016/j.jor.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/19/2024] Open
Abstract
Background Pathogenic degeneration of cartilage and the generation of fibrotic cartilage are crucial characteristics linked to the progression of osteoarthritis (OA). The current research aims to explore the potential function of the miR-137/BMP7 pathway in regulating the fibrogenic transition of chondrocytes associated with OA, as well as assess the therapeutic potential of andrographolide. Methods Samples of cartilage from the knees of patients with OA and individuals without OA were gathered to investigate the expression patterns of miR-137, BMP7, and markers associated with fibrosis. A cell model using primary chondrocytes stimulated with interleukin (IL)-1β was developed to study the involvement of the miR-137/BMP7 axis during the fibrogenic transition of these cells. Additionally, we utilized an animal model of OA in order to assess the beneficial effects of the anti-inflammatory natural compound andrographolide on the fibrogenesis induced by OA in vivo. Results Elevated levels of fibrogenic and inflammatory factors were linked to decreased miR-137 expression in OA samples. In IL-1β-treated chondrocytes, there was an upregulation of fibrogenic markers alongside a reduction in miR-137 levels. The overexpression of miR-137 inhibited fibrogenesis through the negative regulation of BMP7. Additionally, treatment with andrographolide was effective in attenuating the fibrogenic phenotype in chondrocytes and mitigating OA pathogenesis via modulating the miR-137/BMP7 pathway. Conclusion miR-137 downregulation and BMP7 overexpression might contribute to the fibrogenic features in OA-related chondrocytes. Andrographolide attenuates fibrogenic phenotype in chondrocytes and alleviates the severity of OA by modulating the miR-137/BMP7 axis.
Collapse
Affiliation(s)
- Yaoyu Xiang
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Xidan Hu
- Clinical Pharmacy Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Xianguang Yang
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Fei Sun
- Department of Orthopedics, Traditional Chinese Medicine Hospital of Luliang County, Qujing, 655602, Yunnan, China
| | - En Song
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650000, Yunnan, China
| |
Collapse
|
2
|
Huang C, Zeng B, Zhou B, Chen G, Zhang Q, Hou W, Xiao G, Duan L, Hong N, Jin W. Single-cell transcriptomic analysis of chondrocytes in cartilage and pathogenesis of osteoarthritis. Genes Dis 2025; 12:101241. [PMID: 39759119 PMCID: PMC11697194 DOI: 10.1016/j.gendis.2024.101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/26/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2025] Open
Abstract
Chondrocyte is considered the only cell type in cartilage. However, the cell heterogeneity of chondrocytes in human articular cartilage is still not well defined, which hinders our understanding of the pathogenesis of osteoarthritis (OA). Here, we constructed a single-cell transcriptomic atlas of chondrocytes in healthy cartilage and identified nine chondrocyte subsets including homeostatic chondrocytes, proliferate fibrochondrocytes, and hypertrophic chondrocytes (HTC). Interestingly, we identified two distinct HTC subpopulations, among which HTC-1 specifically expressed genes associated with apoptosis and programmed cell death. We identified two main trajectories of chondrocytes, one of which differentiates into fibrochondrocytes, while the other terminates in apoptosis. Comparison of chondrocyte subsets between healthy and OA cartilage showed that proliferate fibrochondrocytes and HTC-1 expanded in OA patients, whereas homeostatic chondrocytes decreased. Interestingly, we discovered an OA-specific proliferate fibrochondrocyte subset that may contribute to the development of OA via inflammation. In summary, this study significantly enhanced our understanding of cell heterogeneity of chondrocytes in articular cartilage and provides insight into the pathogenesis of OA.
Collapse
Affiliation(s)
- Changyuan Huang
- Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bin Zeng
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen, Guangdong 518035, China
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi 53020, China
| | - Bo Zhou
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Guanming Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Qi Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wenhong Hou
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523710, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen, Guangdong 518055, China
| | - Li Duan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University, Shenzhen, Guangdong 518035, China
| | - Ni Hong
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
3
|
Zha Y, Luo X, Ge Z, Zhang J, Li Y, Zhang S. KIAA1199/CEMIP knockdown attenuates cardiac remodeling post myocardial infarction by activating TSP4 pathway in mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167473. [PMID: 39173890 DOI: 10.1016/j.bbadis.2024.167473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Excessive activation of cardiac fibroblasts (CFs) significantly contributes to adverse cardiac remodeling post-myocardial infarction (MI). CEMIP, initially recognized as an enzyme involved in hyaluronic acid (HA) degradation, has also been implicated in the activation of pulmonary fibroblasts. Nevertheless, the role and mechanism of CEMIP in adverse cardiac remodeling following MI remain largely unexplored. MATERIALS AND METHODS RNA sequencing (RNA-seq) was performed on cardiac tissue harvested from the infarct/peri-infarct region of mice 28 days post-MI. RNA-seq was conducted on primary cardiac fibroblasts (CFs) transfected with adenovirus overexpressing CEMIP. Adeno-associated virus serotype 9 (AAV9) was engineered for in vivo CEMIP knockdown to elucidate its impact on cardiac remodeling. Immunoprecipitation coupled with mass spectrometry (IP-MS) and co-immunoprecipitation (co-IP) were employed to elucidate the mechanism by which CEMIP affected cardiac remodeling. KEY FINDINGS RNA-seq of fibrotic heart tissue at day 28 post-MI revealed a significant upregulation of CEMIP. In vitro, CEMIP facilitated the activation of cardiac fibroblasts. In vivo, knockdown of CEMIP markedly reduced cardiac fibrosis and improved cardiac function post-MI. IP-MS and co-immunoprecipitation (co-IP) confirmed that CEMIP interacted with TSP4 through the G8 domain. Further experiments confirmed that CEMIP promoted TSP4 degradation in lysosomes in an ACTN4-dependent manner, thereby activating the FAK signaling pathway. SIGNIFICANCE Our findings suggest that CEMIP significantly contributes to cardiac remodeling post-MI, which might be a novel approach for treating cardiac fibrosis following MI.
Collapse
Affiliation(s)
- Yafang Zha
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Xueyang Luo
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhuowang Ge
- Department of Cardiology, Xinhua School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayan Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Yanyan Li
- Department of Cardiology, Xinhua School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Song Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.
| |
Collapse
|
4
|
Li X, Zhang W, Li X. CEMIP induces TGF-β/Smad signaling to promote keloid development by binding to SPARC. Clinics (Sao Paulo) 2024; 79:100523. [PMID: 39481283 PMCID: PMC11564991 DOI: 10.1016/j.clinsp.2024.100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/08/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Cell Migration Inducing Hyaluronidase 1 (CEMIP) is a protein that plays regulatory functions in a variety of cellular processes in many diseases. Nevertheless, its role and molecular mechanism in keloid hyperplasia are still elusive. METHODS Expressions of CEMIP and Secreted Protein acidic and Rich in Cysteine (SPARC) were detected by qRT-PCR and western blot. CCK-8 assay, along with immunofluorescence staining, was applied for the assessment of cell proliferation. The capabilities of cells to migrate and invade were evaluated utilizing wound healing and Transwell, while Extracellular Matrix (ECM) deposition was measured by immunofluorescence and western blot. The interaction of CEMIP and SPARC was predicted by the Coexpedia and PPA-red databases and verified by co-IP. Western blot was adopted for the estimation of TGF-β/Smad pathway-related proteins. RESULTS The data demonstrated that CEMIP expression was elevated in Keloid Fibroblasts (KF). CEMIP interference suppressed cell proliferative, migrative and invasive capabilities and ECM deposition in KF. Mechanistically, bioinformatics analysis revealed that CEMIP was co-expressed with SPARC and CEMIP protein could bind to SPARC. SPARC expression was reduced in CEMIP-silenced cells. SPARC overexpression counteracted the impacts of CEMIP silencing on cell proliferative, migrative and invasive capabilities and ECM deposition in KF. In addition, the expressions of TGF-β/Smad signaling-related proteins were decreased by CEMIP silencing via the inhibition of SPARC. CONCLUSION In summary, this study revealed that CEMIP modulated KF proliferation, migration, invasion and ECM deposition by TGF-β/Smad signaling through binding to SPARC.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Zhang
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaojing Li
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Zhang Z, Wu W, Li M, Du L, Li J, Yin X, Zhang W. Mesenchymal stem cell–derived extracellular vesicles: A novel nanoimmunoregulatory tool in musculoskeletal diseases. NANO TODAY 2024; 57:102343. [DOI: 10.1016/j.nantod.2024.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Chan DD, Guilak F, Sah RL, Calve S. Mechanobiology of Hyaluronan: Connecting Biomechanics and Bioactivity in Musculoskeletal Tissues. Annu Rev Biomed Eng 2024; 26:25-47. [PMID: 38166186 DOI: 10.1146/annurev-bioeng-073123-120541] [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] [Indexed: 01/04/2024]
Abstract
Hyaluronan (HA) plays well-recognized mechanical and biological roles in articular cartilage and synovial fluid, where it contributes to tissue structure and lubrication. An understanding of how HA contributes to the structure of other musculoskeletal tissues, including muscle, bone, tendon, and intervertebral discs, is growing. In addition, the use of HA-based therapies to restore damaged tissue is becoming more prevalent. Nevertheless, the relationship between biomechanical stimuli and HA synthesis, degradation, and signaling in musculoskeletal tissues remains understudied, limiting the utility of HA in regenerative medicine. In this review, we discuss the various roles and significance of endogenous HA in musculoskeletal tissues. We use what is known and unknown to motivate new lines of inquiry into HA biology within musculoskeletal tissues and in the mechanobiology governing HA metabolism by suggesting questions that remain regarding the relationship and interaction between biological and mechanical roles of HA in musculoskeletal health and disease.
Collapse
Affiliation(s)
- Deva D Chan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA
| | - Robert L Sah
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
7
|
Liu C, Sun Y, Li D, Wang F, Wang H, An S, Sun S. A multifunctional nanogel encapsulating layered double hydroxide for enhanced osteoarthritis treatment via protection of chondrocytes and ECM. Mater Today Bio 2024; 26:101034. [PMID: 38596826 PMCID: PMC11002310 DOI: 10.1016/j.mtbio.2024.101034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/20/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024] Open
Abstract
Osteoarthritis (OA) is characterized by progressive and irreversible damage to the articular cartilage and a consecutive inflammatory response. However, the majority of clinical drugs for OA treatment only alleviate symptoms without addressing the fundamental pathology. To mitigate this issue, we developed an inflammation-responsive carrier and encapsulated bioactive material, namely, LDH@TAGel. The LDH@TAGel was designed with anti-inflammatory and antioxidative abilities, aiming to directly address the pathology of cartilage damage. In particular, LDH was confirmed to restore the ECM secretion function of damaged chondrocytes and attenuate the expression of catabolic matrix metalloproteinases (Mmps). While TAGel showed antioxidant properties by scavenging ROS directly. In vitro evaluation revealed that the LDH@TAGel could protect chondrocytes from inflammation-induced oxidative stress and apoptosis via the Nrf2/Keap1 system and Pi3k-Akt pathway. In vivo experiments demonstrated that the LDH@TAGel could alleviated the degeneration and degradation of cartilage induced by anterior cruciate ligament transection (ACLT). The OARSI scores indicating OA severity decreased significantly after three weeks of intervention. Moreover, the IVIS image revealed that LDH@TAGel enhances the controlled release of LDH in a manner that can be customized according to the severity of OA, allowing adaptive, precise treatment. In summary, this novel design effectively alleviates the underlying pathological causes of OA-related cartilage damage and has emerged as a promising biomaterial for adaptive, cause-targeted OA therapies.
Collapse
Affiliation(s)
- Changxing Liu
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Yawei Sun
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Dengju Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Fan Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Senbo An
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| |
Collapse
|
8
|
Li Y, Huang R, Ye J, Han X, Meng T, Song D, Yin H. Identification of key eRNAs for intervertebral disc degeneration by integrated multinomial bioinformatics analysis. BMC Musculoskelet Disord 2024; 25:356. [PMID: 38704519 PMCID: PMC11069191 DOI: 10.1186/s12891-024-07438-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a common degenerative condition leading to abnormal stress distribution under load, causing intervertebral stenosis, facet joint degeneration, and foraminal stenosis. Very little is known about the molecular mechanism of eRNAs in IVDD. METHODS Gene expression profiles of 38 annulus disc samples composed of 27 less degenerated discs (LDs) and 11 more degenerated discs (MDs) were retrieved from the GEO database. Then, differentially expressed enhancer RNAs (DEeRNAs), differentially expressed target genes (DETGs), and differentially expressed transcription factors (DETFs), hallmark of cancer signalling pathways according to GSVA; the types and quantity of immune cells according to CIBERSORT; and immune gene sets according to ssGSEA were analysed to construct an IVDD-related eRNA network. Then, multidimensional validation was performed to explore the interactions among DEeRNAs, DETFs and DEGs in space. RESULTS A total of 53 components, 14 DETGs, 15 DEeRNAs, 3 DETFs, 5 immune cells, 9 hallmarks, and 7 immune gene sets, were selected to construct the regulatory network. After validation by online multidimensional databases, 21 interactive DEeRNA-DEG-DETF axes related to IVDD exacerbation were identified, among which the C1S-CTNNB1-CHD4 axis was the most significant. CONCLUSION Based upon the results of our study, we theorize that the C1S-CTNNB1-CHD4 axis plays a vital role in IVDD exacerbation. Specifically, C1S recruits CTNNB1 and upregulates the expression of CHD4 in IVDD, and subsequently, CHD4 suppresses glycolysis and activates oxidative phosphorylation, thus generating insoluble collagen fibre deposits and leading to the progression of IVDD. Overall, these DEeRNAs could comprise promising therapeutic targets for IVDD due to their high tissue specificity.
Collapse
Affiliation(s)
- Yongai Li
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianxin Ye
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaying Han
- Department of General Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Meng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Liu Q, Han M, Wu Z, Fu W, Ji J, Liang Q, Tan M, Zhai L, Gao J, Shi D, Jiang Q, Sun Z, Lai Y, Xu Q, Sun Y. DDX5 inhibits hyaline cartilage fibrosis and degradation in osteoarthritis via alternative splicing and G-quadruplex unwinding. NATURE AGING 2024; 4:664-680. [PMID: 38760576 PMCID: PMC11108786 DOI: 10.1038/s43587-024-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/04/2024] [Indexed: 05/19/2024]
Abstract
Hyaline cartilage fibrosis is typically considered an end-stage pathology of osteoarthritis (OA), which results in changes to the extracellular matrix. However, the mechanism behind this is largely unclear. Here, we found that the RNA helicase DDX5 was dramatically downregulated during the progression of OA. DDX5 deficiency increased fibrosis phenotype by upregulating COL1 expression and downregulating COL2 expression. In addition, loss of DDX5 aggravated cartilage degradation by inducing the production of cartilage-degrading enzymes. Chondrocyte-specific deletion of Ddx5 led to more severe cartilage lesions in the mouse OA model. Mechanistically, weakened DDX5 resulted in abundance of the Fn1-AS-WT and Plod2-AS-WT transcripts, which promoted expression of fibrosis-related genes (Col1, Acta2) and extracellular matrix degradation genes (Mmp13, Nos2 and so on), respectively. Additionally, loss of DDX5 prevented the unfolding Col2 promoter G-quadruplex, thereby reducing COL2 production. Together, our data suggest that strategies aimed at the upregulation of DDX5 hold significant potential for the treatment of cartilage fibrosis and degradation in OA.
Collapse
Affiliation(s)
- Qianqian Liu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Mingrui Han
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhigui Wu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wenqiang Fu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, China
| | - Jun Ji
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qingqing Liang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Dongquan Shi
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ziying Sun
- Department of Orthopaedics, Jinling Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuping Lai
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
10
|
Guo S, Guo Y, Chen Y, Cui S, Zhang C, Chen D. The role of CEMIP in cancers and its transcriptional and post-transcriptional regulation. PeerJ 2024; 12:e16930. [PMID: 38390387 PMCID: PMC10883155 DOI: 10.7717/peerj.16930] [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: 11/15/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
CEMIP is a protein known for inducing cell migration and binding to hyaluronic acid. Functioning as a hyaluronidase, CEMIP primarily facilitates the breakdown of the extracellular matrix component, hyaluronic acid, thereby regulating various signaling pathways. Recent evidence has highlighted the significant role of CEMIP in different cancers, associating it with diverse pathological states. While identified as a biomarker for several diseases, CEMIP's mechanism in cancer seems distinct. Accumulating data suggests that CEMIP expression is triggered by chemical modifications to itself and other influencing factors. Transcriptionally, chemical alterations to the CEMIP promoter and involvement of transcription factors such as AP-1, HIF, and NF-κB regulate CEMIP levels. Similarly, specific miRNAs have been found to post-transcriptionally regulate CEMIP. This review provides a comprehensive summary of CEMIP's role in various cancers and explores how both transcriptional and post-transcriptional mechanisms control its expression.
Collapse
Affiliation(s)
- Song Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yunfei Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yuanyuan Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Shuaishuai Cui
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Chunmei Zhang
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Dahu Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| |
Collapse
|
11
|
Kuchynsky K, Stevens P, Hite A, Xie W, Diop K, Tang S, Pietrzak M, Khan S, Walter B, Purmessur D. Transcriptional profiling of human cartilage endplate cells identifies novel genes and cell clusters underlying degenerated and non-degenerated phenotypes. Arthritis Res Ther 2024; 26:12. [PMID: 38173036 PMCID: PMC10763221 DOI: 10.1186/s13075-023-03220-6] [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: 08/30/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Low back pain is a leading cause of disability worldwide and is frequently attributed to intervertebral disc (IVD) degeneration. Though the contributions of the adjacent cartilage endplates (CEP) to IVD degeneration are well documented, the phenotype and functions of the resident CEP cells are critically understudied. To better characterize CEP cell phenotype and possible mechanisms of CEP degeneration, bulk and single-cell RNA sequencing of non-degenerated and degenerated CEP cells were performed. METHODS Human lumbar CEP cells from degenerated (Thompson grade ≥ 4) and non-degenerated (Thompson grade ≤ 2) discs were expanded for bulk (N=4 non-degenerated, N=4 degenerated) and single-cell (N=1 non-degenerated, N=1 degenerated) RNA sequencing. Genes identified from bulk RNA sequencing were categorized by function and their expression in non-degenerated and degenerated CEP cells were compared. A PubMed literature review was also performed to determine which genes were previously identified and studied in the CEP, IVD, and other cartilaginous tissues. For single-cell RNA sequencing, different cell clusters were resolved using unsupervised clustering and functional annotation. Differential gene expression analysis and Gene Ontology, respectively, were used to compare gene expression and functional enrichment between cell clusters, as well as between non-degenerated and degenerated CEP samples. RESULTS Bulk RNA sequencing revealed 38 genes were significantly upregulated and 15 genes were significantly downregulated in degenerated CEP cells relative to non-degenerated cells (|fold change| ≥ 1.5). Of these, only 2 genes were previously studied in CEP cells, and 31 were previously studied in the IVD and other cartilaginous tissues. Single-cell RNA sequencing revealed 11 unique cell clusters, including multiple chondrocyte and progenitor subpopulations with distinct gene expression and functional profiles. Analysis of genes in the bulk RNA sequencing dataset showed that progenitor cell clusters from both samples were enriched in "non-degenerated" genes but not "degenerated" genes. For both bulk- and single-cell analyses, gene expression and pathway enrichment analyses highlighted several pathways that may regulate CEP degeneration, including transcriptional regulation, translational regulation, intracellular transport, and mitochondrial dysfunction. CONCLUSIONS This thorough analysis using RNA sequencing methods highlighted numerous differences between non-degenerated and degenerated CEP cells, the phenotypic heterogeneity of CEP cells, and several pathways of interest that may be relevant in CEP degeneration.
Collapse
Affiliation(s)
- Kyle Kuchynsky
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Patrick Stevens
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amy Hite
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - William Xie
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA
| | - Khady Diop
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Shirley Tang
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Safdar Khan
- Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Benjamin Walter
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA
| | - Devina Purmessur
- Department of Biomedical Engineering, The Ohio State University, 3016 Fontana Laboratories, 140 W. 19th Ave, Columbus, OH, 43210, USA.
- Department of Orthopaedics, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
12
|
Zheng S, An S, Luo Y, Vithran DTA, Yang S, Lu B, Deng Z, Li Y. HYBID in osteoarthritis: Potential target for disease progression. Biomed Pharmacother 2023; 165:115043. [PMID: 37364478 DOI: 10.1016/j.biopha.2023.115043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
HYBID is a new hyaluronan-degrading enzyme and exists in various cells of the human body. Recently, HYBID was found to over-express in the osteoarthritic chondrocytes and fibroblast-like synoviocytes. According to these researches, high level of HYBID is significantly correlated with cartilage degeneration in joints and hyaluronic acid degradation in synovial fluid. In addition, HYBID can affect inflammatory cytokine secretion, cartilage and synovium fibrosis, synovial hyperplasia via multiple signaling pathways, thereby exacerbating osteoarthritis. Based on the existing research of HYBID in osteoarthritis, HYBID can break the metabolic balance of HA in joints through the degradation ability independent of HYALs/CD44 system and furthermore affect cartilage structure and mechanotransduction of chondrocytes. In particular, in addition to HYBID itself being able to trigger some signaling pathways, we believe that low-molecular-weight hyaluronan produced by excess degradation can also stimulate some disease-promoting signaling pathways by replacing high-molecular-weight hyaluronan in joints. The specific role of HYBID in osteoarthritis is gradually revealed, and the discovery of HYBID raises the new way to treat osteoarthritis. In this review, the expression and basic functions of HYBID in joints were summarized, and reveal potential role of HYBID as a key target in treatment for osteoarthritis.
Collapse
Affiliation(s)
- Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Senbo An
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Luo
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Djandan Tadum Arthur Vithran
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shaoqu Yang
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
13
|
Faber BG, Frysz M, Boer CG, Evans DS, Ebsim R, Flynn KA, Lundberg M, Southam L, Hartley A, Saunders FR, Lindner C, Gregory JS, Aspden RM, Lane NE, Harvey NC, Evans DM, Zeggini E, Davey Smith G, Cootes T, Van Meurs J, Kemp JP, Tobias JH. The identification of distinct protective and susceptibility mechanisms for hip osteoarthritis: findings from a genome-wide association study meta-analysis of minimum joint space width and Mendelian randomisation cluster analyses. EBioMedicine 2023; 95:104759. [PMID: 37619450 PMCID: PMC10470292 DOI: 10.1016/j.ebiom.2023.104759] [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: 05/26/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Hip minimum joint space width (mJSW) provides a proxy for cartilage thickness. This study aimed to conduct a genome-wide association study (GWAS) of mJSW to (i) identify new genetic determinants of mJSW and (ii) identify which mJSW loci convey hip osteoarthritis (HOA) risk and would therefore be of therapeutic interest. METHODS GWAS meta-analysis of hip mJSW derived from plain X-rays and DXA was performed, stratified by sex and adjusted for age and ancestry principal components. Mendelian randomisation (MR) and cluster analyses were used to examine causal effect of mJSW on HOA. FINDINGS 50,745 individuals were included in the meta-analysis. 42 SNPs, which mapped to 39 loci, were identified. Mendelian randomisation (MR) revealed little evidence of a causal effect of mJSW on HOA (ORIVW 0.98 [95% CI 0.82-1.18]). However, MR-Clust analysis suggested the null MR estimates reflected the net effect of two distinct causal mechanisms cancelling each other out, one of which was protective, whereas the other increased HOA susceptibility. For the latter mechanism, all loci were positively associated with height, suggesting mechanisms leading to greater height and mJSW increase the risk of HOA in later life. INTERPRETATIONS One group of mJSW loci reduce HOA risk via increased mJSW, suggesting possible utility as targets for chondroprotective therapies. The second group of mJSW loci increased HOA risk, despite increasing mJSW, but were also positively related to height, suggesting they contribute to HOA risk via a growth-related mechanism. FUNDING Primarily funded by the Medical Research Council and Wellcome Trust.
Collapse
Affiliation(s)
- Benjamin G Faber
- Musculoskeletal Research Unit, University of Bristol, UK; Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK.
| | - Monika Frysz
- Musculoskeletal Research Unit, University of Bristol, UK; Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK
| | - Cindy G Boer
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, USA
| | - Raja Ebsim
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, UK
| | - Kaitlyn A Flynn
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia
| | - Mischa Lundberg
- UQ Frazer Institute, The University of Queensland, Woolloongabba, Australia
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Centre for Environmental Health, Neuherberg, Germany
| | - April Hartley
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK
| | - Fiona R Saunders
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, UK
| | - Claudia Lindner
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, UK
| | - Jennifer S Gregory
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, UK
| | - Richard M Aspden
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, UK
| | - Nancy E Lane
- Center for Musculoskeletal Health, University of California Davis, Sacramento, USA
| | - Nicholas C Harvey
- Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, UK
| | - David M Evans
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK; Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia; UQ Frazer Institute, The University of Queensland, Woolloongabba, Australia
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Centre for Environmental Health, Neuherberg, Germany; Technical University of Munich and Klinikum Rechts der Isar, TUM School of Medicine, Germany
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK
| | - Timothy Cootes
- Division of Informatics, Imaging and Data Sciences, The University of Manchester, UK
| | - Joyce Van Meurs
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - John P Kemp
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK; Mater Research Institute, The University of Queensland, Woolloongabba, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia
| | - Jonathan H Tobias
- Musculoskeletal Research Unit, University of Bristol, UK; Medical Research Council Integrative Epidemiology Unit at the University of Bristol, UK
| |
Collapse
|
14
|
Liu Y, Hu G, Li Y, Kong X, Yang K, Li Z, Lao W, Li J, Zhong J, Zhang S, Leng Y, Bi C, Zhai A. Research on the biological mechanism and potential application of CEMIP. Front Immunol 2023; 14:1222425. [PMID: 37662915 PMCID: PMC10471826 DOI: 10.3389/fimmu.2023.1222425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
Cell migration-inducing protein (CEMIP), also known as KIAA1199 and hyaluronan-binding protein involved in hyaluronan depolymerization, is a new member of the hyaluronidase family that degrades hyaluronic acid (HA) and remodels the extracellular matrix. In recent years, some studies have reported that CEMIP can promote the proliferation, invasion, and adhesion of various tumor cells and can play an important role in bacterial infection and arthritis. This review focuses on the pathological mechanism of CEMIP in a variety of diseases and expounds the function of CEMIP from the aspects of inhibiting cell apoptosis, promoting HA degradation, inducing inflammatory responses and related phosphorylation, adjusting cellular microenvironment, and regulating tissue fibrosis. The diagnosis and treatment strategies targeting CEMIP are also summarized. The various functions of CEMIP show its great potential application value.
Collapse
Affiliation(s)
- Yang Liu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Gang Hu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuetong Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinyi Kong
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Kaming Yang
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhenlin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wanwen Lao
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiaxin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jianhua Zhong
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shitong Zhang
- Department of General Practice, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuxin Leng
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Changlong Bi
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Aixia Zhai
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
15
|
Spataro S, Guerra C, Cavalli A, Sgrignani J, Sleeman J, Poulain L, Boland A, Scapozza L, Moll S, Prunotto M. CEMIP (HYBID, KIAA1199): structure, function and expression in health and disease. FEBS J 2023; 290:3946-3962. [PMID: 35997767 DOI: 10.1111/febs.16600] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022]
Abstract
CEMIP (cell migration-inducing protein), also known as KIAA1199 or HYBID, is a protein involved in the depolymerisation of hyaluronic acid (HA), a major glycosaminoglycan component of the extracellular matrix. CEMIP was originally described in patients affected by nonsyndromic hearing loss and has subsequently been shown to play a key role in tumour initiation and progression, as well as arthritis, atherosclerosis and idiopathic pulmonary fibrosis. Despite the vast literature associating CEMIP with these diseases, its biology remains elusive. The present review article summarises all the major scientific evidence regarding its structure, function, role and expression, and attempts to cast light on a protein that modulates EMT, fibrosis and tissue inflammation, an unmet key aspect in several inflammatory disease conditions.
Collapse
Affiliation(s)
- Sofia Spataro
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Concetta Guerra
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Jonathan Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute for Biological and Chemical Systems - Biological Information Processing (IBCS - BIP), Karlsruhe Institute for Technology (KIT), Germany
| | - Lina Poulain
- Department of Molecular Biology, University of Geneva, Switzerland
| | - Andreas Boland
- Department of Molecular Biology, University of Geneva, Switzerland
| | - Leonardo Scapozza
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Solange Moll
- Department of Pathology, University Hospital of Geneva, Switzerland
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| |
Collapse
|
16
|
Fusi G, Constantinides M, Fissoun C, Pichard L, Pers YM, Ferreira-Lopez R, Pantesco V, Poulet C, Malaise O, De Seny D, Lemaitre JM, Jorgensen C, Brondello JM. Senescence-Driven Inflammatory and Trophic Microenvironment Imprints Mesenchymal Stromal/Stem Cells in Osteoarthritic Patients. Biomedicines 2023; 11:1994. [PMID: 37509633 PMCID: PMC10377055 DOI: 10.3390/biomedicines11071994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Senescent cells promote progressive tissue degeneration through the establishment of a combined inflammatory and trophic microenvironment. The cellular senescence state has therefore emerged as a central driving mechanism of numerous age-related diseases, including osteoarthritis (OA), the most common rheumatic disease. Senescence hallmarks are detectable in chondrocytes, synoviocytes and sub-chondral bone cells. This study investigates how the senescence-driven microenvironment could impact the cell fate of resident osteoarticular mesenchymal stromal/stem cells (MSCs) that are hence contributing to OA disease progression. For that purpose, we performed a comparative gene expression analysis of MSCs isolated from healthy donors that were in vitro chronically exposed either to interferon-gamma (IFN-γ) or Transforming Growth Factor beta 1 (TGFβ1), two archetypical factors produced by senescent cells. Both treatments reduced MSC self-renewal capacities by upregulating different senescence-driven cycle-dependent kinase inhibitors. Furthermore, a common set of differentially expressed genes was identified in both treated MSCs that was also found enriched in MSCs isolated from OA patients. These findings highlight an imprinting of OA MSCs by the senescent joint microenvironment that changes their matrisome gene expression. Altogether, this research gives new insights into OA etiology and points to new innovative therapeutic opportunities to treat OA patients.
Collapse
Affiliation(s)
- Giuseppe Fusi
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
| | | | | | - Lydiane Pichard
- SAFE-iPSC Facility INGESTEM, Montpellier University Hospital, 34298 Montpellier, France
| | - Yves-Marie Pers
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier University Hospital, 34298 Montpellier, France
| | - Rosanna Ferreira-Lopez
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier University Hospital, 34298 Montpellier, France
| | | | - Christophe Poulet
- Laboratory and Service of Rheumatology, GIGA-I3, Université de Liège, 4000 Liege, Belgium
| | - Olivier Malaise
- Laboratory and Service of Rheumatology, GIGA-I3, Université de Liège, 4000 Liege, Belgium
| | - Dominique De Seny
- Laboratory and Service of Rheumatology, GIGA-I3, Université de Liège, 4000 Liege, Belgium
| | - Jean-Marc Lemaitre
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- SAFE-iPSC Facility INGESTEM, Montpellier University Hospital, 34298 Montpellier, France
| | - Christian Jorgensen
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier University Hospital, 34298 Montpellier, France
| | | |
Collapse
|
17
|
Zhang S, Wang P, Hu B, Lv X, Liu W, Chen S, Shao Z. Inhibiting Heat Shock Protein 90 Attenuates Nucleus Pulposus Fibrosis and Pathologic Angiogenesis Induced by Macrophages via Down-Regulating Cell Migration-Inducing Protein. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:960-976. [PMID: 37088454 DOI: 10.1016/j.ajpath.2023.03.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 04/25/2023]
Abstract
Intervertebral disc (IVD) degeneration (IVDD) is usually accompanied by nucleus pulposus (NP) fibrosis and pathologic angiogenesis, which are possibly associated with macrophage infiltration. Previous research indicates a destructive role of macrophages and the protective effect of inhibiting heat shock protein 90 (HSP90) in IVDD. Herein, the effects of inhibiting HSP90 on NP fibrosis and pathologic angiogenesis induced by macrophages were investigated further. Single-cell RNA-sequencing analysis was used to classify fibrotic NP cell (NPC) clusters and healthy NPC clusters in human NP tissues. The fibrotic NPC clusters were possibly associated with angiogenesis-related biological processes. Immunostaining showed the spatial association between blood vessel ingrowth and macrophage infiltration, as well as elevated levels of cell migration-inducing protein (CEMIP) and vascular endothelial growth factor A in severely degenerated human IVD tissues. Particularly, HSP90 inhibitor tanespimycin (17-AAG) ameliorated macrophage-induced fibrotic phenotype of NPCs via inhibiting CEMIP. M2, but not M1, macrophages promoted the pro-angiogenic ability of endothelial cells, which was attenuated by 17-AAG or HSP90 siRNA. Reversing the fibrotic phenotype of NPCs by Cemip siRNA also mitigated the pro-angiogenic effects of M2-conditioned medium-treated NPCs. Moreover, the murine IVDD model supported the 17-AAG-induced amelioration of NP fibrosis and endothelial cell invasion in IVD tissues. In conclusion, inhibiting HSP90 attenuated two interrelated pathologic processes, NP fibrosis and pathologic angiogenesis, induced by macrophages via down-regulating CEMIP.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Liu M, Xie L, Zhang Y, Chen J, Zhang X, Chen Y, Huang W, Cai M, Liang L, Lai M, Huang J, Guo Y, Lin L, Zhu K. Inhibition of CEMIP potentiates the effect of sorafenib on metastatic hepatocellular carcinoma by reducing the stiffness of lung metastases. Cell Death Dis 2023; 14:25. [PMID: 36639658 PMCID: PMC9839779 DOI: 10.1038/s41419-023-05550-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Hepatocellular carcinoma (HCC) with lung metastasis is associated with poor prognosis and poor therapeutic outcomes. Studies have demonstrated that stiffened stroma can promote metastasis in various tumors. However, how the lung mechanical microenvironment favors circulating tumor cells remains unclear in metastatic HCC. Here, we found that the expression of cell migration-inducing hyaluronan-binding protein (CEMIP) was closely associated with lung metastasis and can promote pre-metastatic niche formation by increasing lung matrix stiffness. Furthermore, upregulated serum CEMIP was indicative of lung fibrotic changes severity in patients with HCC lung metastasis. By directly targeting CEMIP, pirfenidone can inhibit CEMIP/TGF-β1/Smad signaling pathway and reduce lung metastases stiffening, demonstrating promising antitumor activity. Pirfenidone in combination with sorafenib can more effectively suppress the incidence of lung metastasis compared with sorafenib alone. This study is the first attempt to modulate the mechanical microenvironment for HCC therapy and highlights CEMIP as a potential target for the prevention and treatment of HCC lung metastasis. CEMIP mediating an HCC-permissive microenvironment through controlling matrix stiffness. Meanwhile, Pirfenidone could reduce metastasis stiffness and increases the anti-angiogenic effect of Sorafenib by directly targeting CEMIP.
Collapse
Affiliation(s)
- Mingyu Liu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Lulu Xie
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Yuying Zhang
- Central Laboratory, Shenzhen Longhua Maternity and Child Healthcare Hospital, 518109, Shenzhen, China
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, 510630, Guangzhou, China
| | - Xiang Zhang
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease and The Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Chen
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Wensou Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Mingyue Cai
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Licong Liang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Miaoling Lai
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Jingjun Huang
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Yongjian Guo
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| | - Liteng Lin
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| | - Kangshun Zhu
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, and Department of Radiology, the Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Hua Q, Lu Y, Wang D, Da J, Peng W, Sun G, Gu K, Wang H, Zhu Y. KIAA1199 promotes oxaliplatin resistance and epithelial mesenchymal transition of colorectal cancer via protein O-GlcNAcylation. Transl Oncol 2023; 28:101617. [PMID: 36610242 PMCID: PMC9850197 DOI: 10.1016/j.tranon.2023.101617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Oxaliplatin is a commonly used platinum drug for colorectal cancer (CRC). However, the treatment of CRC by oxaliplatin usually fails because of drug resistance, which results in a huge challenge in the therapy of CRC. Elucidation of molecular mechanisms may help to overcome oxaliplatin resistance of CRC. In our study, we revealed that KIAA1199 can promote oxaliplatin resistance of CRC. Mechanistically, KIAA1199 prevents oxaliplatin mediated apoptosis via up-regulated PARP1 derived from reduced endoplasmic reticulum stress induced by protein O-GlcNAcylation. In the meantime, KIAA1199 can also trigger epithelial mesenchymal transition by stabilizing SNAI1 protein via O-GlcNAcylation. Therefore, KIAA1199 has great potential to be a novel biomarker, therapeutic target for oxaliplatin resistance and metastasis of CRC.
Collapse
Affiliation(s)
- Qingling Hua
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yuanyuan Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu 241004, China
| | - Dingxiang Wang
- Department of Psychology, The fourth people's hospital, Wuhu, 241003, China
| | - Jie Da
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Wanren Peng
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China
| | - Yanzhe Zhu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei 230022, China,Corresponding author.
| |
Collapse
|
20
|
Direct comparison of non-osteoarthritic and osteoarthritic synovial fluid-induced intracellular chondrocyte signaling and phenotype changes. Osteoarthritis Cartilage 2023; 31:60-71. [PMID: 36150677 DOI: 10.1016/j.joca.2022.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Since the joint microenvironment and tissue homeostasis are highly dependent on synovial fluid, we aimed to compare the essential chondrocyte signaling signatures of non-osteoarthritic vs end-stage osteoarthritic knee synovial fluid. Moreover, we determined the phenotypic consequence of the distinct signaling patterns on articular chondrocytes. METHODS Protein profiling of synovial fluid was performed using antibody arrays. Chondrocyte signaling and phenotypic changes induced by non-osteoarthritic and osteoarthritic synovial fluid were analyzed using a phospho-kinase array, luciferase-based transcription factor activity assays, and RT-qPCR. The origin of osteoarthritic synovial fluid signaling was evaluated by comparing the signaling responses of conditioned media from cartilage, synovium, infrapatellar fat pad and meniscus. Osteoarthritic synovial fluid induced pathway-phenotype relationships were evaluated using pharmacological inhibitors. RESULTS Compared to non-osteoarthritic synovial fluid, osteoarthritic synovial fluid was enriched in cytokines, chemokines and growth factors that provoked differential MAPK, AKT, NFκB and cell cycle signaling in chondrocytes. Functional pathway analysis confirmed increased activity of these signaling events upon osteoarthritic synovial fluid stimulation. Tissue secretomes of osteoarthritic cartilage, synovium, infrapatellar fat pad and meniscus activated several inflammatory signaling routes. Furthermore, the distinct pathway signatures of osteoarthritic synovial fluid led to accelerated chondrocyte dedifferentiation via MAPK/ERK signaling, increased chondrocyte fibrosis through MAPK/JNK and PI3K/AKT activation, an elevated inflammatory response mediated by cPKC/NFκB, production of extracellular matrix-degrading enzymes by MAPK/p38 and PI3K/AKT routes, and enabling of chondrocyte proliferation. CONCLUSION This study provides the first mechanistic comparison between non-osteoarthritic and osteoarthritic synovial fluid, highlighting MAPKs, cPKC/NFκB and PI3K/AKT as crucial OA-associated intracellular signaling routes.
Collapse
|
21
|
Plaas AHK, Moran MM, Sandy JD, Hascall VC. Aggrecan and Hyaluronan: The Infamous Cartilage Polyelectrolytes - Then and Now. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:3-29. [PMID: 37052843 DOI: 10.1007/978-3-031-25588-5_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Cartilages are unique in the family of connective tissues in that they contain a high concentration of the glycosaminoglycans, chondroitin sulfate and keratan sulfate attached to the core protein of the proteoglycan, aggrecan. Multiple aggrecan molecules are organized in the extracellular matrix via a domain-specific molecular interaction with hyaluronan and a link protein, and these high molecular weight aggregates are immobilized within the collagen and glycoprotein network. The high negative charge density of glycosaminoglycans provides hydrophilicity, high osmotic swelling pressure and conformational flexibility, which together function to absorb fluctuations in biomechanical stresses on cartilage during movement of an articular joint. We have summarized information on the history and current knowledge obtained by biochemical and genetic approaches, on cell-mediated regulation of aggrecan metabolism and its role in skeletal development, growth as well as during the development of joint disease. In addition, we describe the pathways for hyaluronan metabolism, with particular focus on the role as a "metabolic rheostat" during chondrocyte responses in cartilage remodeling in growth and disease.Future advances in effective therapeutic targeting of cartilage loss during osteoarthritic diseases of the joint as an organ as well as in cartilage tissue engineering would benefit from 'big data' approaches and bioinformatics, to uncover novel feed-forward and feed-back mechanisms for regulating transcription and translation of genes and their integration into cell-specific pathways.
Collapse
Affiliation(s)
- Anna H K Plaas
- Department of Internal Medicine (Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | - Meghan M Moran
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - John D Sandy
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Vincent C Hascall
- Department of Biomedical Engineering, The Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
22
|
Liu J, Xia S, Han P, Zhang M, Wu J, Liao J. Downregulation of KIAA1199 alleviated the activation, proliferation, and migration of hepatic stellate cells by the inhibition of epithelial-mesenchymal transition. Open Med (Wars) 2023; 18:20230689. [PMID: 37034499 PMCID: PMC10080707 DOI: 10.1515/med-2023-0689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
KIAA1199, a major glycosaminoglycan component of the extracellular matrix, was reported to induce a fibrosis-like process. However, the relationship between KIAA1199 and liver fibrosis remains unclear. The liver fibrosis mouse model was established with carbon tetrachloride (CCl4). Here, we found that KIAA1199 was upregulated in CCl4-induced liver fibrosis. The expression of KIAA1199 was also increased in TGF-β-stimulated LX-2 cells. To clarify the impact of KIAA1199 in hepatic stellate cells (HSCs), we downregulated the expression of KIAA1199 in LX-2 cells by RNA interference. Cell proliferation, apoptosis, and migration were determined by CCK-8, flow cytometry, and transwell assay. We found that KIAA1199 knockdown reduced the expression of fibrosis markers α-SMA and COL1A1. Depletion of KIAA1199 inhibited cell proliferation by downregulating cyclin B1 and cyclin D1 and promoted cell apoptosis by upregulating Bax and downregulating Bcl-2. Moreover, KIAA1199 knockdown decreased matrix metalloproteinase-2 (MMP-2) and MMP-9 expression to inhibit the migration ability of LX-2 cells. Silencing KIAA1199 also suppressed the epithelial-mesenchymal transition phenomenon. Collectively, our study revealed that KIAA1199 knockdown alleviated the activation, proliferation, and migration of HSCs, while promoting apoptosis of HSCs, which suggests that KIAA1199 may be a potential regulator of liver fibrosis.
Collapse
Affiliation(s)
- Jingmei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingyu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jingwen Wu
- Department of Gastroenterology, Hainan Hospital of PLA General Hospital, Sanya, Hainan, China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
23
|
Wang C, Liu Y, Zhang W, Huang J, Jiang J, Wang R, Zeng D. circ-BPTF serves as a miR-486-5p sponge to regulate CEMIP and promotes hypoxic pulmonary arterial smooth muscle cell proliferation in COPD. Acta Biochim Biophys Sin (Shanghai) 2022; 55:438-448. [PMID: 36514216 PMCID: PMC10160238 DOI: 10.3724/abbs.2022178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Hypoxia plays a crucial role in pulmonary vascular remodelling at the early stage of chronic obstructive pulmonary disease (COPD). Circle RNA (circRNA) has been identified to play a critical role in multiple diseases. However, the role of circRNAs in pulmonary vascular remodelling in COPD remains unclear. In this study, we aim to investigate the role of circRNAs in pulmonary arterial smooth muscle cell proliferation and pulmonary vascular remodelling in COPD. COPD patients show lower partial pressure of arterial oxygen and pulmonary arterial remodeling as compared with controls. circRNA microarray and real-time PCR analyses show significantly higher level of circ-BPTF and lower miR-486-5p level in the pulmonary arteries of COPD patients as compared with controls. Hypoxia suppresses miR-486-5p expression but promotes expressions of circ-BPTF and cell migration inducing protein (CEMIP) in human pulmonary arterial smooth muscle cells (PASMCs) in vitro. Loss- and gain-of-function experiments show that circ-BPTF promotes PASMC proliferation in vitro. Moreover, luciferase reporter assay results indicate that circ-BPTF regulates PASMC proliferation by acting as an miR-486-5p sponge. CEMIP is identified as a candidate target gene of miR-486-5p by luciferase reporter assay. Overall, our study shows that circ-BPTF serves as a miR-486-5p sponge to regulate CEMIP and promote hypoxic PASMC proliferation in pulmonary vascular remodelling in COPD.
Collapse
Affiliation(s)
- Changguo Wang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yingying Liu
- Department of Pulmonary and Critical Care Medicine, Suzhou Dushu Lake Hospital, Suzhou 215006, China.,Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215006, China
| | - Weiyun Zhang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Department of Pulmonary and Critical Care Medicine, Suzhou Dushu Lake Hospital, Suzhou 215006, China.,Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215006, China
| | - Jian'an Huang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Junhong Jiang
- Department of Pulmonary and Critical Care Medicine, Suzhou Dushu Lake Hospital, Suzhou 215006, China.,Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215006, China
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Daxiong Zeng
- Department of Pulmonary and Critical Care Medicine, Suzhou Dushu Lake Hospital, Suzhou 215006, China.,Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215006, China
| |
Collapse
|
24
|
Loss of the repressor REST affects progesterone receptor function and promotes uterine leiomyoma pathogenesis. Proc Natl Acad Sci U S A 2022; 119:e2205524119. [PMID: 36282915 PMCID: PMC9636955 DOI: 10.1073/pnas.2205524119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Uterine leiomyomas (UL) are benign tumors that arise in the myometrial layer of the uterus. The standard treatment option for UL is hysterectomy, although hormonal therapies, such as selective progesterone receptor modulators, are often used as temporary treatment options to reduce symptoms or to slow the growth of tumors. However, since the pathogenesis of UL is poorly understood and most hormonal therapies are not based on UL-specific, divergent hormone signaling pathways, hallmarks that predict long-term efficacy and safety of pharmacotherapies remain largely undefined. In a previous study, we reported that aberrant expression of repressor element 1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) target genes activate UL growth due to the near ubiquitous loss of REST. Here, we show that ablation of the Rest gene in mouse uterus leads to UL phenotype and gene-expression patterns analogous to UL, including altered estrogen and progesterone signaling pathways. We demonstrate that many of the genes dysregulated in UL harbor cis-regulatory elements bound by REST and progesterone receptor (PGR) adjacent to each other. Crucially, we identify an interaction between REST and PGR in healthy myometrium and present a putative mechanism for the dysregulation of progesterone-responsive genes in UL ensuing in the loss of REST. Using three Rest conditional knockout mouse lines, we provide a comprehensive picture of the impact loss of REST has in UL pathogenesis and in altering the response of UL to steroid hormones.
Collapse
|
25
|
Domanegg K, Sleeman JP, Schmaus A. CEMIP, a Promising Biomarker That Promotes the Progression and Metastasis of Colorectal and Other Types of Cancer. Cancers (Basel) 2022; 14:cancers14205093. [PMID: 36291875 PMCID: PMC9600181 DOI: 10.3390/cancers14205093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary CEMIP (cell migration-inducing and hyaluronan-binding protein) has been implicated in the pathogenesis of numerous diseases, including colorectal and other forms of cancer. The molecular functions of CEMIP are currently under investigation and include the degradation of the extracellular matrix component hyaluronic acid (HA), as well as the regulation of a number of signaling pathways. In this review, we survey our current understanding of how CEMIP contributes to tumor growth and metastasis, focusing particularly on colorectal cancer, for which it serves as a promising biomarker. Abstract Originally discovered as a hypothetical protein with unknown function, CEMIP (cell migration-inducing and hyaluronan-binding protein) has been implicated in the pathogenesis of numerous diseases, including deafness, arthritis, atherosclerosis, idiopathic pulmonary fibrosis, and cancer. Although a comprehensive definition of its molecular functions is still in progress, major functions ascribed to CEMIP include the depolymerization of the extracellular matrix component hyaluronic acid (HA) and the regulation of a number of signaling pathways. CEMIP is a promising biomarker for colorectal cancer. Its expression is associated with poor prognosis for patients suffering from colorectal and other types of cancer and functionally contributes to tumor progression and metastasis. Here, we review our current understanding of how CEMIP is able to foster the process of tumor growth and metastasis, focusing particularly on colorectal cancer. Studies in cancer cells suggest that CEMIP exerts its pro-tumorigenic and pro-metastatic activities through stimulating migration and invasion, suppressing cell death and promoting survival, degrading HA, regulating pro-metastatic signaling pathways, inducing the epithelial–mesenchymal transition (EMT) program, and contributing to the metabolic reprogramming and pre-metastatic conditioning of future metastatic microenvironments. There is also increasing evidence indicating that CEMIP may be expressed in cells within the tumor microenvironment that promote tumorigenesis and metastasis formation, although this remains in an early stage of investigation. CEMIP expression and activity can be therapeutically targeted at a number of levels, and preliminary findings in animal models show encouraging results in terms of reduced tumor growth and metastasis, as well as combating therapy resistance. Taken together, CEMIP represents an exciting new player in the progression of colorectal and other types of cancer that holds promise as a therapeutic target and biomarker.
Collapse
Affiliation(s)
- Kevin Domanegg
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leopoldshafen, Germany
- Correspondence:
| | - Anja Schmaus
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
26
|
Yamamoto K, Scavenius C, Meschis MM, Gremida AME, Mogensen EH, Thøgersen IB, Bonelli S, Scilabra SD, Jensen A, Santamaria S, Ahnström J, Bou-Gharios G, Enghild JJ, Nagase H. A top-down approach to uncover the hidden ligandome of low-density lipoprotein receptor-related protein 1 in cartilage. Matrix Biol 2022; 112:190-218. [PMID: 36028175 DOI: 10.1016/j.matbio.2022.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022]
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a cell-surface receptor ubiquitously expressed in various tissues. It plays tissue-specific roles by mediating endocytosis of a diverse range of extracellular molecules. Dysregulation of LRP1 is involved in multiple conditions including osteoarthritis (OA) but little information is available about the specific profile of direct binding partners of LRP1 (ligandome) for each tissue, which would lead to a better understanding of its role in disease states. Here, we investigated adult articular cartilage where impaired LRP1-mediated endocytosis leads to tissue destruction. We used a top-down approach involving proteomic analysis of the LRP1 interactome in human chondrocytes, direct binding assays using purified LRP1 and ligand candidates, and validation in LRP1-deficient fibroblasts and human chondrocytes, as well as a novel Lrp1 conditional knockout (KO) mouse model. We found that inhibition of LRP1 and ligand interaction results in cell death, alteration of the entire secretome and transcriptional modulations in human chondrocytes. We identified a chondrocyte-specific LRP1 ligandome consisting of more than 50 novel ligand candidates. Surprisingly, 23 previously reported LRP1 ligands were not regulated by LRP1-mediated endocytosis in human chondrocytes. We confirmed direct LRP1 binding of HGFAC, HMGB1, HMGB2, CEMIP, SLIT2, ADAMTS1, TSG6, IGFBP7, SPARC and LIF, correlation between their affinity for LRP1 and the rate of endocytosis, and some of their intracellular localization. Moreover, a conditional LRP1 KO mouse model demonstrated a critical role of LRP1 in regulating the high-affinity ligands in cartilage in vivo. This systematic approach revealed the specificity and the extent of the chondrocyte LRP1 ligandome and identified potential novel therapeutic targets for OA.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom.
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Maria M Meschis
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Abdulrahman M E Gremida
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Emilie H Mogensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Ida B Thøgersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Simone Bonelli
- Fondazione RiMED - ISMETT via Ernesto Tricomi 5, 90127 Palermo, Italy
| | - Simone D Scilabra
- Fondazione RiMED - ISMETT via Ernesto Tricomi 5, 90127 Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Salvatore Santamaria
- Department of Immunology and Inflammation, Imperial College London, Du Cane Road, W12 0NN, London, United Kingdom
| | - Josefin Ahnström
- Department of Immunology and Inflammation, Imperial College London, Du Cane Road, W12 0NN, London, United Kingdom
| | - George Bou-Gharios
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Hideaki Nagase
- Kennedy Institute of Rheumatology, University of Oxford, Headington, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
27
|
Huang X, Lan Y, Shen J, Chen Z, Xie Z. Extracellular Vesicles in Bone Homeostasis: Emerging Mediators of Osteoimmune Interactions and Promising Therapeutic Targets. Int J Biol Sci 2022; 18:4088-4100. [PMID: 35844790 PMCID: PMC9274499 DOI: 10.7150/ijbs.69816] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
An imbalance in bone homeostasis results in bone loss and poor healing in bone diseases and trauma. Osteoimmune interactions, as a key contributor to bone homeostasis, depend on the crosstalk between mesenchymal stem cell-osteoblast (MSC-OB) and monocyte-macrophage (MC-Mφ) lineages. Currently, extracellular vesicles (EVs) are considered to be involved in cell-to-cell communication and represent a novel avenue to enhance our understanding of bone homeostasis and to develop novel diagnostic and therapeutic options. In this comprehensive review, we aim to present recent advances in the study of the effect of MC-Mφ-derived EVs on osteogenesis and the regulatory effects of MSC-OB-derived EVs on the differentiation, recruitment and efferocytosis of Mφ. Furthermore, we discuss the role of EVs as crucial mediators of the communication between these cell lineages involved in the development of common bone diseases, with a focus on osteoporosis, osteoarthritis, bone fracture, and periodontal disease. Together, this review focuses on the apparent discrepancies in current research findings and future directions for translating fundamental insights into clinically relevant EV-based therapies for improving bone health.
Collapse
Affiliation(s)
- Xiaoyuan Huang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yanhua Lan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Jiahui Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Zhuo Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
28
|
Housmans BAC, Neefjes M, Surtel DAM, Vitík M, Cremers A, van Rhijn LW, van der Kraan PM, van den Akker GGH, Welting TJM. Synovial fluid from end-stage osteoarthritis induces proliferation and fibrosis of articular chondrocytes via MAPK and RhoGTPase signaling. Osteoarthritis Cartilage 2022; 30:862-874. [PMID: 35176481 DOI: 10.1016/j.joca.2021.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Alterations in the composition of synovial fluid have been associated with adverse effects on cartilage integrity and function. Here, we examined the phenotypic and proliferative behavior of human articular chondrocytes when cultured in vitro for 13 days with synovial fluid derived from end-stage osteoarthritis patients. MATERIALS AND METHODS Chondrocyte proliferation and phenotypical changes induced by osteoarthritic synovial fluid were analyzed using DNA staining, RT-qPCR, immunostainings, and immunoblotting. The molecular mechanisms by which osteoarthritic synovial fluid induced fibrosis and proliferation were studied using a phospho-protein antibody array and luciferase-based transcription factor activity assays. Specific pathway inhibitors were used to probe the involvement of pathways in fibrosis and proliferation. RESULTS Prolonged stimulation with osteoarthritic synovial fluid sustained chondrocyte proliferation and induced profound phenotypic changes, favoring a fibrotic over a chondrogenic or hypertrophic phenotype. A clear loss of chondrogenic markers at both the transcriptional and protein level was observed, while expression of several fibrosis-associated markers were upregulated over time. Phospho-kinase analysis revealed activation of MAPK and RhoGTPase signaling pathways by osteoarthritic synovial fluid, which was confirmed by elevated transcriptional activity of Elk-1 and SRF. Inhibitor studies revealed that ERK played a central role in the loss of chondrocyte phenotype, while EGFR and downstream mediators p38, JNK and Rac/Cdc42 were essential for fibrosis-associated collagen expression. Finally, we identified EGF signaling as a key activator of chondrocyte proliferation. CONCLUSIONS Osteoarthritic synovial fluid promoted chondrocyte fibrosis and proliferation through EGF receptor activation and downstream MAPK and RhoGTPase signaling.
Collapse
Affiliation(s)
- B A C Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Neefjes
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - D A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - M Vitík
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - A Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - L W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands
| | - T J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Maastricht, the Netherlands; Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
29
|
Deroyer C, Poulet C, Paulissen G, Ciregia F, Malaise O, Plener Z, Cobraiville G, Daniel C, Gillet P, Malaise MG, de Seny D. CEMIP (KIAA1199) regulates inflammation, hyperplasia and fibrosis in osteoarthritis synovial membrane. Cell Mol Life Sci 2022; 79:260. [PMID: 35474501 PMCID: PMC9042994 DOI: 10.1007/s00018-022-04282-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022]
Abstract
Osteoarthritis (OA) synovial membrane is mainly characterized by low-grade inflammation, hyperplasia with increased cell proliferation and fibrosis. We previously underscored a critical role for CEMIP in fibrosis of OA cartilage. However, its role in OA synovial membrane remains unknown. An in vitro model with fibroblast-like synoviocytes from OA patients and an in vivo model with collagenase-induced OA mice were used to evaluate CEMIP-silencing effects on inflammation, hyperplasia and fibrosis. Our results showed that i. CEMIP expression was increased in human and mouse inflamed synovial membrane; ii. CEMIP regulated the inflammatory response pathway and inflammatory cytokines production in vitro and in vivo; iii. CEMIP induced epithelial to mesenchymal transition pathway and fibrotic markers in vitro and in vivo; iv. CEMIP increased cell proliferation and synovial hyperplasia; v. CEMIP expression was increased by inflammatory cytokines and by TGF-β signaling; vi. anti-fibrotic drugs decreased CEMIP expression. All these findings highlighted the central role of CEMIP in OA synovial membrane development and underscored that targeting CEMIP could be a new therapeutic approach.
Collapse
Affiliation(s)
- Céline Deroyer
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium.
| | - Christophe Poulet
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Geneviève Paulissen
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Federica Ciregia
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Olivier Malaise
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Zelda Plener
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Gaël Cobraiville
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | | | - Philippe Gillet
- Department of Orthopaedic Surgery, CHULiège, 4000, Liège, Belgium
| | - Michel G Malaise
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA-Research, CHULiège, ULiège, 4000, Liège, Belgium
| |
Collapse
|
30
|
Lierova A, Kasparova J, Filipova A, Cizkova J, Pekarova L, Korecka L, Mannova N, Bilkova Z, Sinkorova Z. Hyaluronic Acid: Known for Almost a Century, but Still in Vogue. Pharmaceutics 2022; 14:838. [PMID: 35456670 PMCID: PMC9029726 DOI: 10.3390/pharmaceutics14040838] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
Hyaluronic acid (HA) has a special position among glycosaminoglycans. As a major component of the extracellular matrix (ECM). This simple, unbranched polysaccharide is involved in the regulation of various biological cell processes, whether under physiological conditions or in cases of cell damage. This review summarizes the history of this molecule's study, its distinctive metabolic pathway in the body, its unique properties, and current information regarding its interaction partners. Our main goal, however, is to intensively investigate whether this relatively simple polymer may find applications in protecting against ionizing radiation (IR) or for therapy in cases of radiation-induced damage. After exposure to IR, acute and belated damage develops in each tissue depending upon the dose received and the cellular composition of a given organ. A common feature of all organ damage is a distinct change in composition and structure of the ECM. In particular, the important role of HA was shown in lung tissue and the variability of this flexible molecule in the complex mechanism of radiation-induced lung injuries. Moreover, HA is also involved in intermediating cell behavior during morphogenesis and in tissue repair during inflammation, injury, and would healing. The possibility of using the HA polymer to affect or treat radiation tissue damage may point to the missing gaps in the responsible mechanisms in the onset of this disease. Therefore, in this article, we will also focus on obtaining answers from current knowledge and the results of studies as to whether hyaluronic acid can also find application in radiation science.
Collapse
Affiliation(s)
- Anna Lierova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic; (A.F.); (J.C.); (L.P.); (Z.S.)
| | - Jitka Kasparova
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 532 10 Pardubice, Czech Republic; (J.K.); (L.K.); (N.M.); (Z.B.)
| | - Alzbeta Filipova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic; (A.F.); (J.C.); (L.P.); (Z.S.)
| | - Jana Cizkova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic; (A.F.); (J.C.); (L.P.); (Z.S.)
| | - Lenka Pekarova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic; (A.F.); (J.C.); (L.P.); (Z.S.)
| | - Lucie Korecka
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 532 10 Pardubice, Czech Republic; (J.K.); (L.K.); (N.M.); (Z.B.)
| | - Nikola Mannova
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 532 10 Pardubice, Czech Republic; (J.K.); (L.K.); (N.M.); (Z.B.)
| | - Zuzana Bilkova
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, 532 10 Pardubice, Czech Republic; (J.K.); (L.K.); (N.M.); (Z.B.)
| | - Zuzana Sinkorova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, 500 01 Hradec Kralove, Czech Republic; (A.F.); (J.C.); (L.P.); (Z.S.)
| |
Collapse
|
31
|
Possible Repositioning of an Oral Anti-Osteoporotic Drug, Ipriflavone, for Treatment of Inflammatory Arthritis via Inhibitory Activity of KIAA1199, a Novel Potent Hyaluronidase. Int J Mol Sci 2022; 23:ijms23084089. [PMID: 35456905 PMCID: PMC9030858 DOI: 10.3390/ijms23084089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
KIAA1199 has a strong hyaluronidase activity in inflammatory arthritis. This study aimed to identify a drug that could reduce KIAA1199 activity and clarify its effects on inflammatory arthritis. Rat chondrosarcoma (RCS) cells were strongly stained with Alcian blue (AB). Its stainability was reduced in RCS cells, which were over-expressed with the KIAA1199 gene (RCS-KIAA). We screened the drugs that restore the AB stainability in RCS-KIAA. The effects of the drug were evaluated by particle exclusion assay, HA ELISA, RT-PCR, and Western blotting. We further evaluated the HA accumulation and the MMP1 and three expressions in fibroblast-like synoviocytes (FLS). In vivo, the effects of the drug on symptoms and serum concentration of HA in a collagen-induced arthritis mouse were evaluated. Ipriflavone was identified to restore AB stainability at 23%. Extracellular matrix formation was significantly increased in a dose-dependent manner (p = 0.006). Ipriflavone increased the HA accumulation and suppressed the MMP1 and MMP3 expression on TNF-α stimulated FLS. In vivo, Ipriflavone significantly improved the symptoms and reduced the serum concentrations of HA. Conclusions: We identified Ipriflavone, which has inhibitory effects on KIAA1199 activity. Ipriflavone may be a therapeutic candidate based on its reduction of KIAA1199 activity in inflammatory arthritis.
Collapse
|
32
|
Thielen NGM, Neefjes M, Vitters EL, van Beuningen HM, Blom AB, Koenders MI, van Lent PLEM, van de Loo FAJ, Blaney Davidson EN, van Caam APM, van der Kraan PM. Identification of Transcription Factors Responsible for a Transforming Growth Factor-β-Driven Hypertrophy-like Phenotype in Human Osteoarthritic Chondrocytes. Cells 2022; 11:cells11071232. [PMID: 35406794 PMCID: PMC8998018 DOI: 10.3390/cells11071232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022] Open
Abstract
During osteoarthritis (OA), hypertrophy-like chondrocytes contribute to the disease process. TGF-β's signaling pathways can contribute to a hypertrophy(-like) phenotype in chondrocytes, especially at high doses of TGF-β. In this study, we examine which transcription factors (TFs) are activated and involved in TGF-β-dependent induction of a hypertrophy-like phenotype in human OA chondrocytes. We found that TGF-β, at levels found in synovial fluid in OA patients, induces hypertrophic differentiation, as characterized by increased expression of RUNX2, COL10A1, COL1A1, VEGFA and IHH. Using luciferase-based TF activity assays, we observed that the expression of these hypertrophy genes positively correlated to SMAD3:4, STAT3 and AP1 activity. Blocking these TFs using specific inhibitors for ALK-5-induced SMAD signaling (5 µM SB-505124), JAK-STAT signaling (1 µM Tofacitinib) and JNK signaling (10 µM SP-600125) led to the striking observation that only SB-505124 repressed the expression of hypertrophy factors in TGF-β-stimulated chondrocytes. Therefore, we conclude that ALK5 kinase activity is essential for TGF-β-induced expression of crucial hypertrophy factors in chondrocytes.
Collapse
|
33
|
Schmaus A, Rothley M, Schreiber C, Möller S, Roßwag S, Franz S, Garvalov BK, Thiele W, Spataro S, Herskind C, Prunotto M, Anderegg U, Schnabelrauch M, Sleeman J. Sulfated hyaluronic acid inhibits the hyaluronidase CEMIP and regulates the HA metabolism, proliferation and differentiation of fibroblasts. Matrix Biol 2022; 109:173-191. [DOI: 10.1016/j.matbio.2022.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/12/2022] [Accepted: 04/04/2022] [Indexed: 12/23/2022]
|
34
|
Bolia IK, Mertz K, Faye E, Sheppard J, Telang S, Bogdanov J, Hasan LK, Haratian A, Evseenko D, Weber AE, Petrigliano FA. Cross-Communication Between Knee Osteoarthritis and Fibrosis: Molecular Pathways and Key Molecules. Open Access J Sports Med 2022; 13:1-15. [PMID: 35261547 PMCID: PMC8898188 DOI: 10.2147/oajsm.s321139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/18/2022] [Indexed: 01/26/2023] Open
Abstract
Knee fibrosis is characterized by the presence of excessive connective tissue due to dysregulated fibroblast activation following local or systemic tissue damage. Knee fibrosis constitutes a major clinical problem in orthopaedics due to the severe limitation in the knee range of motion that leads to compromised function and patient disability. Knee osteoarthritis is an extremely common orthopedic condition that is associated with patient disability and major costs to the health-care systems worldwide. Although knee fibrosis and osteoarthritis (OA) have traditionally been perceived as two separate pathologic entities, recent research has shown common ground between the pathophysiologic processes that lead to the development of these two conditions. The purpose of this review was to identify the pathophysiologic pathways as well as key molecules that are implicated in the development of both knee OA and knee fibrosis in order to understand the relationship between the two diagnoses and potentially identify novel therapeutic targets.
Collapse
Affiliation(s)
- Ioanna K Bolia
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA,Correspondence: Ioanna K Bolia, 1520 San Pablo Street Suite 2000, Los Angeles, CA, 90033, USA, Tel +1 9703432813, Fax +1 818-658-5925, Email
| | - Kevin Mertz
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Ethan Faye
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Justin Sheppard
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Sagar Telang
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Jacob Bogdanov
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Laith K Hasan
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Aryan Haratian
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Denis Evseenko
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Alexander E Weber
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| | - Frank A Petrigliano
- USC Epstein Family Center for Sports Medicine at Keck Medicine of USC, Los Angeles, CA, USA
| |
Collapse
|
35
|
Chen CH, Ke GM, Lin PC, Lin KD. Therapeutic DNA vaccine encoding CEMIP (KIAA1199) ameliorates kidney fibrosis in obesity through inhibiting the Wnt/β-catenin pathway. Biochim Biophys Acta Gen Subj 2021; 1865:130019. [PMID: 34582938 DOI: 10.1016/j.bbagen.2021.130019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND CEMIP is a novel risk factor of various cancers through activating Wnt/β-catenin /epithelial-mesenchymal transition between epithelial cells and stroma. The chronic fibrosis commonly contributes renal carcinogenesis in patients with obesity. As there have very few choices of medicines targeting CEMIP. This study intended to design therapeutic DNA vaccines for nephropathy in obesity, through diminishing the CEMIP/Wnt1/β-catenin pathway. METHOD In an 8-week experiment, plasmid-encoding CEMIP was vaccinated into high-fat diet (HFD) or obesity mice in the first 4 weeks, and then vaccination was stopped for at least 4 weeks. Then, plasma and spleens were harvested to evaluate anti-CEMIP antibody synthesis and T-helper type 1 and 2 activation after vaccination. Kidneys were collected to investigate efficacy of CEMIP DNA vaccine on inhibiting HFD and obesity-induced fibrosis and Wnt1/β-catenin pathway. To confirm that CEMIP crucially contributed towards fibrotic formation, CEMIP gene or siRNA transfection was performed in HK-2 cells under VLDL stimulation, or not. RESULTS At the end point, anti-CEMIP antibody was successfully produced in the pcDNA 3.1-CEMIP vaccinated group, while Wnt1/β-catenin signaling and fibrosis was inactive. Through VLDL stimulation and CEMIP overexpression, Wnt1/β-catenin signaling and fibrosis significantly presented in vitro. Otherwise, anti-sera of CEMIP-vaccinated mice could inhibit the VLDL-induced Wnt1/β-catenin/fibrosis pathway in HK-2 cells. Similarly, the silencing of CEMIP by siRNA ameliorated the Wnt1/β-catenin pathway and fibrogenesis under VLDL stimulation. CONCLUSION DNA vaccine targeting CEMIP/Wnt1/β-catenin pathway plays a novel strategy in nephropathy. GENERAL SIGNIFICANCE Immune therapy might provide a new therapeutic option on nephropathy of obesity.
Collapse
Affiliation(s)
- Chao-Hung Chen
- General Research Service Center, National Pingtung University of Science and Technology, Pingtung, Taiwan; Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan; Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Guan-Ming Ke
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Pi-Chen Lin
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kun-Der Lin
- Division of Endocrinology and Metabolism, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan..
| |
Collapse
|
36
|
Malaise O, Paulissen G, Deroyer C, Ciregia F, Poulet C, Neuville S, Plener Z, Daniel C, Gillet P, Lechanteur C, Brondello JM, de Seny D, Malaise M. Influence of Glucocorticoids on Cellular Senescence Hallmarks in Osteoarthritic Fibroblast-like Synoviocytes. J Clin Med 2021; 10:jcm10225331. [PMID: 34830613 PMCID: PMC8617749 DOI: 10.3390/jcm10225331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/25/2021] [Accepted: 11/13/2021] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is recognized as being a cellular senescence-linked disease. Intra-articular injections of glucocorticoids (GC) are frequently used in knee OA to treat synovial effusion but face controversies about toxicity. We investigated the influence of GC on cellular senescence hallmarks and senescence induction in fibroblast-like synoviocytes (FLS) from OA patients and mesenchymal stem cells (MSC). Methods: Cellular senescence was assessed via the proliferation rate, β-galactosidase staining, DNA damage and CKI expression (p21, p16INK4A). Experimental senescence was induced by irradiation. Results: The GC prednisolone did not induce an apparent senescence phenotype in FLS, with even higher proliferation, no accumulation of β-galactosidase-positive cells nor DNA damage and reduction in p21mRNA, only showing the enhancement of p16INK4A. Prednisolone did not modify experimental senescence induction in FLS, with no modulation of any senescence parameters. Moreover, prednisolone did not induce a senescence phenotype in MSC: despite high β-galactosidase-positive cells, no reduction in proliferation, no DNA damage and no CKI enhancement was observed. Conclusions: We provide reassuring in vitro data about the use of GC regarding cellular senescence involvement in OA: the GC prednisolone did not induce a senescent phenotype in OA FLS (the proliferation ratio was even higher) and in MSC and did not worsen cellular senescence establishment.
Collapse
Affiliation(s)
- Olivier Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
- Correspondence: ; Tel.: +32-4-366-7863
| | - Geneviève Paulissen
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Céline Deroyer
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Federica Ciregia
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Christophe Poulet
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Sophie Neuville
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Zelda Plener
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Christophe Daniel
- Orthopedic Surgery Department, CHU de Liège, 4000 Liège, Belgium; (C.D.); (P.G.)
| | - Philippe Gillet
- Orthopedic Surgery Department, CHU de Liège, 4000 Liège, Belgium; (C.D.); (P.G.)
| | - Chantal Lechanteur
- Laboratory of Cell and Gene Therapy, Department of Hematology, CHU de Liège, 4000 Liège, Belgium;
| | - Jean-Marc Brondello
- Institute for Regenerative Medicine and Biotherapy, Univ Montpellier, INSERM UMR1183, 34298 Montpellier, France;
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| | - Michel Malaise
- Laboratory of Rheumatology, GIGA Research, CHU de Liège, University of Liège, 4000 Liège, Belgium; (G.P.); (C.D.); (F.C.); (C.P.); (S.N.); (Z.P.); (D.d.S.); (M.M.)
| |
Collapse
|
37
|
Electroacupuncture Upregulates HIF-1 α and SOX9 Expression in Knee Osteoarthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2047097. [PMID: 34760015 PMCID: PMC8575628 DOI: 10.1155/2021/2047097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 01/13/2023]
Abstract
Electroacupuncture (EA) has been clinically used in knee osteoarthritis broadly and proved to be effective than other therapies with fewer side effects; however, the mechanism of electroacupuncture to work on cartilage remains unclear. In this study, we aimed to evaluate the effect of EA treatment on cartilage and the relationship between EA and proteins such as HIF-a and SOX9. EA (dilatational wave, 3-15 HZ, 1 mA) has been applied to bilateral Zusanli (ST36), Xuehai (SP10), Taixi (KI3), and Yanglingquan (GB34) of rats. Results showed that the cartilage of the knee osteoarthritis group had obvious damage and fissure formation while the EA group showed that the cartilage destruction was generally milder. In addition, the protein expression levels of HIF-1α, and chondrogenic markers such as Sox9, and ACAN in the electroacupuncture group were higher than those in the ACLT group. Also, the extracellular matrix protein expression levels of MMP13 and ADAMTS5 were decreased in the EA group. These findings indicate that EA could alleviate the severity of knee osteoarthritis, and HIF-a and SOX9 may closely attribute to the treatment.
Collapse
|
38
|
Abstract
The fibrocartilage chondrocyte phenotype has been recognized to attribute to osteoarthritis (OA) development. These chondrocytes express genes related to unfavorable OA outcomes, emphasizing its importance in OA pathology. BMP7 is being explored as a potential disease-modifying molecule and attenuates the chondrocyte hypertrophic phenotype. On the other hand, BMP7 has been demonstrated to relieve organ fibrosis by counteracting the pro-fibrotic TGFβ-Smad3-PAI1 axis and increasing MMP2-mediated Collagen type I turnover. Whether BMP7 has anti-fibrotic properties in chondrocytes is unknown. Human OA articular chondrocytes (HACs) were isolated from end-stage OA femoral cartilage (total knee arthroplasty; n = 18 individual donors). SW1353 cells and OA HACs were exposed to 1 nM BMP7 for 24 h, after which gene expression of fibrosis-related genes and fibrosis-mediating factors was determined by RT-qPCR. In SW1353, Collagen type I protein levels were determined by immunocytochemistry and western blotting. PAI1 and MMP2 protein levels and activity were measured with an ELISA and activity assays, respectively. MMP2 activity was inhibited with the selective MMP-2 inhibitor OA-Hy. SMAD3 activity was determined by a (CAGA)12-reporter assay, and pSMAD2 levels by western blotting. Following BMP7 exposure, the expression of fibrosis-related genes was reduced in SW1353 cells and OA HACs. BMP7 reduced Collagen type I protein levels in SW1353 cells. Gene expression of MMP2 was increased in SW1353 cells following BMP7 treatment. BMP7 reduced PAI1 protein levels and -activity, while MMP2 protein levels and -activity were increased by BMP7. BMP7-dependent inhibition of Collagen type I protein levels in SW1353 cells was abrogated when MMP2 activity was inhibited. Finally, BMP7 reduced pSMAD2 levels determined by western blotting and reduced SMAD3 transcriptional activity as demonstrated by decreased (CAGA)12 luciferase reporter activity. Our data demonstrate that short-term exposure to BMP7 decreases the fibrocartilage chondrocyte phenotype. The BMP7-dependent reduction of Collagen type I protein expression seems MMP2-dependent and inhibition of Smad2/3-PAI1 activity was identified as a potential pathway via which BMP7 exerts its anti-fibrotic action. This indicates that in chondrocytes BMP7 may have a double mode-of-action by targeting both the hypertrophic as well as the fibrotic chondrocyte phenotype, potentially adding to the clinical relevance of using BMP7 as an OA disease-modifying molecule.
Collapse
|
39
|
Sun JS, Wang WW, Lian HK. The Clinical Significance of Changes in Serum New Cytokine CYTL1 in Patients with Knee Osteoarthritis. Int J Gen Med 2021; 14:5105-5109. [PMID: 34511992 PMCID: PMC8420777 DOI: 10.2147/ijgm.s322370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/16/2021] [Indexed: 01/28/2023] Open
Abstract
Objective The present study aims to investigate the clinical significance of changes in the expression of new cytokine-like 1 (CYTL1) in the serum of patients with knee osteoarthritis (KOA). Methods A total of 182 patients with KOA, including 84 males and 98 females aged 39–86 with an average age of 66.4 ± 9.7 and an average body mass index (BMI) of 24.9 ± 2.4 kg/m2, were enrolled in the study. The patients were divided into three subgroups: the grade II subgroup (n = 23), grade III subgroup (n = 63), and grade IV subgroup (n = 96) based on severity, as calculated by the Kellgren and Lawrence (K&L) classification system. In addition, 152 volunteers from our health center who came in for physical examination were selected as the control group, including 70 males and 82 females aged 37–82 with an average age of 63.4 ± 9.5 and an average BMI of 24.8 ± 2.2 kg/m2. An enzyme-linked immunosorbent assay was adopted to detect the serum CYTL1 levels, and the correlation between CYTL1 and the severity of KOA was analyzed. Results The serum level of CYTL1 was significantly lower in the KOA group than in the control group (P < 0.05). In the KOA group, the difference in the serum level of CYTL1 was statistically significant between the subgroups and decreased significantly with an increase in the severity of the disease (F = 54.826, P < 0.001). Therefore, the serum level of CYTL1 was correlated with the severity of the disease, as determined by the K&L classification system (r = −0.613, P < 0.001). Conclusion The serum levels of CYTL1 are strongly correlated with the severity of the disease in patients with KOA and could be a new therapeutic target for KOA.
Collapse
Affiliation(s)
- Jian-Sheng Sun
- Department of Orthopedics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Wei-Wei Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Hong-Kai Lian
- Department of Orthopedics, Zhengzhou Central Hospital, Zhengzhou, 450000, Henan, People's Republic of China
| |
Collapse
|
40
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China ,grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Peng Liu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
41
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z+10.1038/s41413-021-00163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2024] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
42
|
Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
|
43
|
Momoeda M, de Vega S, Kaneko H, Yoshinaga C, Shimoda M, Nakamura T, Endo Y, Yoshida H, Kaneko K, Ishijima M, Okada Y. Deletion of Hybid (Hyaluronan-Binding Protein Involved in Hyaluronan Depolymerization) Results in Attenuation of Osteoarthritis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1986-1998. [PMID: 34390681 DOI: 10.1016/j.ajpath.2021.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/29/2021] [Accepted: 07/28/2021] [Indexed: 10/20/2022]
Abstract
Hyaluronan (HA)-binding protein involved in HA depolymerization (HYBID) is involved in cartilage destruction via HA depolymerization in human knee osteoarthritis. However, roles of HYBID in the progression of osteoarthritis remain elusive. This study sought to examine whether genetic depletion of Hybid could suppress surgically induced osteoarthritis of mouse knee joints. In osteoarthritis induced by medial collateral ligament transection with meniscus removal, articular cartilage destruction and osteophyte formation at the medial femoral-tibial joint were significantly inhibited in Hybid-deficient (Hybid-/-) mouse group compared with wild-type group. Hybid was highly produced by synovial cells and articular chondrocytes in wild-type mouse osteoarthritis joint. IL-1β, IL-6, and tumor necrosis factor-α were up-regulated in the osteoarthritis joint tissues of both wild-type and Hybid-/- mice. Vascular density at the synovial and periosteal junction was significantly reduced in Hybid-/- mice compared with wild-type mice. High-molecular-weight HA was accumulated in osteoarthritis joint tissues of Hybid-/- mice. Injections of high-molecular-weight HA to knee joints attenuated the cartilage destruction and osteophyte formation in wild-type mouse osteoarthritis group. Inhibition of cartilage destruction and osteophyte formation in Hybid-/- mice was also observed in destabilization of the medial meniscus model. These data are the first to demonstrate that cartilage destruction and osteophyte formation are suppressed in Hybid-/- mice and suggest that Hybid-mediated HA depolymerization is implicated for the progression of mechanically induced knee osteoarthritis.
Collapse
Affiliation(s)
- Masahiro Momoeda
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Susana de Vega
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Haruka Kaneko
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chiho Yoshinaga
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Tomomi Nakamura
- Biological Science Research, Kao Corporation, Kanagawa, Japan
| | - Yoko Endo
- Biological Science Research, Kao Corporation, Kanagawa, Japan
| | | | - Kazuo Kaneko
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
44
|
Guo H, Yang J, Liu S, Qin T, Zhao Q, Hou X, Ren L. Prognostic marker identification based on weighted gene co-expression network analysis and associated in vitro confirmation in gastric cancer. Bioengineered 2021; 12:4666-4680. [PMID: 34338150 PMCID: PMC8806585 DOI: 10.1080/21655979.2021.1957645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The aim of this study was to explore the potential molecular mechanisms of Gastric cancer (GC) and identify new prognostic markers for GC. RNA sequencing data were downloaded from the Gene Expression Omnibus database, and 418 differentially expressed genes (DEGs) were screened. Weighted correlation network analysis (WGCNA) was performed to identify six hub modules related to the clinical features of GC. Cytoscape software was used to identify five hub genes in the co-expression network, including CST1, CEMIP, COL8A1, PMEPA1, and MSLN. The TCGA database was used to verify hub gene expression in GC. The overall survival in the high CEMIP expression group was significantly lower than that of patients in the low CEMIP expression group. CEMIP expression was also found to be negatively correlated with B cell and CD4 + T cell infiltration. Further, associated in vitro experiments confirmed that CEMIP downregulation suppressed the proliferation and migration of GC cells and impaired the chemoresistance of GC cells to 5-fluorouracil. Our study effectively identified and validated prognostic biomarkers for GC, laying a new foundation for the therapeutic target, occurrence, and development of gastric cancer.
Collapse
Affiliation(s)
- Haonan Guo
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Jun Yang
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Shanshan Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Tao Qin
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Qianwen Zhao
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Xianliang Hou
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Lei Ren
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| |
Collapse
|
45
|
Tan Y, Luo X, Lv W, Hu W, Zhao C, Xiong M, Yi Y, Wang D, Wang Y, Wang H, Wu Y, Zhang Q. Tumor-derived exosomal components: the multifaceted roles and mechanisms in breast cancer metastasis. Cell Death Dis 2021; 12:547. [PMID: 34039961 PMCID: PMC8155106 DOI: 10.1038/s41419-021-03825-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 02/04/2023]
Abstract
Breast cancer (BC) is the most frequently invasive malignancy and the leading cause of tumor-related mortality among women worldwide. Cancer metastasis is a complex, multistage process, which eventually causes tumor cells to colonize and grow at the metastatic site. Distant organ metastases are the major obstacles to the management of advanced BC patients. Notably, exosomes are defined as specialized membrane-enclosed extracellular vesicles with specific biomarkers, which are found in a wide variety of body fluids. Recent studies have demonstrated that exosomes are essential mediators in shaping the tumor microenvironment and BC metastasis. The transferred tumor-derived exosomes modify the capability of invasive behavior and organ-specific metastasis in recipient cells. BC exosomal components, mainly including noncoding RNAs (ncRNAs), proteins, lipids, are the most investigated components in BC metastasis. In this review, we have emphasized the multifaceted roles and mechanisms of tumor-derived exosomes in BC metastasis based on these important components. The underlying mechanisms mainly include the invasion behavior change, tumor vascularization, the disruption of the vascular barrier, and the colonization of the targeted organ. Understanding the significance of tumor-derived exosomal components in BC metastasis is critical for yielding novel routes of BC intervention.
Collapse
Affiliation(s)
- Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Xiao Luo
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Wenchang Lv
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Weijie Hu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Chongru Zhao
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Mingchen Xiong
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yi Yi
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Dawei Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Yichen Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Haiping Wang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| | - Yiping Wu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| |
Collapse
|
46
|
Modulation of α Vβ 6 integrin in osteoarthritis-related synovitis and the interaction with VTN (381-397 a.a.) competing for TGF-β1 activation. Exp Mol Med 2021; 53:210-222. [PMID: 33526813 PMCID: PMC8080589 DOI: 10.1038/s12276-021-00558-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis is characterized by structural alteration of joints. Fibrosis of the synovial tissue is often detected and considered one of the main causes of joint stiffness and pain. In our earlier proteomic study, increased levels of vitronectin (VTN) fragment (amino acids 381–397) were observed in the serum of osteoarthritis patients. In this work, the affinity of this fragment for integrins and its putative role in TGF-β1 activation were investigated. A competition study determined the interaction of VTN(381–397 a.a.) with αVβ6 integrin. Subsequently, the presence of αVβ6 integrin was substantiated on primary human fibroblast-like synoviocytes (FLSs) by western blot and flow cytometry. By immunohistochemistry, β6 was detected in synovial membranes, and its expression showed a correlation with tissue fibrosis. Moreover, β6 expression was increased under TGF-β1 stimulation; hence, a TGF-β bioassay was applied. We observed that αVβ6 could mediate TGF-β1 bioavailability and that VTN(381–397 a.a.) could prevent TGF-β1 activation by interacting with αVβ6 in human FLSs and increased α-SMA. Finally, we analyzed serum samples from healthy controls and patients with osteoarthritis and other rheumatic diseases by nano-LC/Chip MS–MS, confirming the increased expression of VTN(381–397 a.a.) in osteoarthritis as well as in lupus erythematosus and systemic sclerosis. These findings corroborate our previous observations concerning the overexpression of VTN(381–397 a.a.) in osteoarthritis but also in other rheumatic diseases. This fragment interacts with αVβ6 integrin, a receptor whose expression is increased in FLSs from the osteoarthritic synovial membrane and that can mediate the activation of the TGF-β1 precursor in human FLSs. Insights into a mechanism underlying the formation of fibrotic tissue within joints in osteoarthritis may also prove relevant to other rheumatological disorders. The general mechanisms underlying fibrosis are reasonably well understood, but it remains unclear what triggers these processes in osteoarthitis. Researchers of the University of Liège in Belgium have uncovered a possible explanation based on experiments with cultured primary synovial fibroblasts from patients. Osteoarthitis is characterized by increased levels of a fragment of the protein vitronectin, and the researchers demonstrated that this in turn binds to a protein called αVβ6, potentially promoting initiation of fibrosis. They also observed elevated levels of the same vitronectin fragment in two other rheumatoid disorders, lupus and systemic sclerosis, and concluded that further research is needed to characterize this protein’s role in inflammation and fibrosis.
Collapse
|
47
|
Rim YA, Ju JH. The Role of Fibrosis in Osteoarthritis Progression. Life (Basel) 2020; 11:life11010003. [PMID: 33374529 PMCID: PMC7822172 DOI: 10.3390/life11010003] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease where the main characteristics include cartilage degeneration and synovial membrane inflammation. These changes in the knee joint eventually dampen the function of the joint and restrict joint movement, which eventually leads to a stage where total joint replacement is the only treatment option. While much is still unknown about the pathogenesis and progression mechanism of OA, joint fibrosis can be a critical issue for better understanding this disease. Synovial fibrosis and the generation of fibrocartilage are the two main fibrosis-related characteristics that can be found in OA. However, these two processes remain mostly misunderstood. In this review, we focus on the fibrosis process in OA, especially in the cartilage and the synovium tissue, which are the main tissues involved in OA.
Collapse
Affiliation(s)
- Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-6895
| |
Collapse
|
48
|
Charlier E, Deroyer C, Neuville S, Plener Z, Malaise O, Ciregia F, Gillet P, Reuter G, Salvé M, Withofs N, Hustinx R, de Seny D, Malaise MG. Toward diagnostic relevance of the α Vβ 5, α Vβ 3, and α Vβ 6 integrins in OA: expression within human cartilage and spinal osteophytes. Bone Res 2020; 8:35. [PMID: 33083095 PMCID: PMC7527564 DOI: 10.1038/s41413-020-00110-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/06/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
We previously reported 18FPRGD2 uptake by the coxofemoral lining, intervertebral discs and facet joint osteophytes in OA using PET/SCAN imaging. However, the molecular mechanism by which the PRGD2 tracer interacts with joint tissues and osteophytes in OA remains unclear. As PRGD2 ligands are expected to belong to the RGD-specific integrin family, the purpose of this study was (i) to determine which integrin complexes display the highest affinity for PRGD2-based ligands, (ii) to analyze integrin expression in relevant tissues, and (iii) to test integrin regulation in chondrocytes using OA-related stimuli to increase the levels of fibrosis and ossification markers. To this end, the affinity of PRGD2-based ligands for five heterodimeric integrins was measured by competition with 125I-echistatin. In situ analyses were performed in human normal vs. OA cartilage and spinal osteophytes. Osteophytes were characterized by (immuno-)histological staining. Integrin subunit expression was tested in chondrocytes undergoing dedifferentiation, osteogenic differentiation, and inflammatory stimulation. The integrins αVβ5, αVβ3, and αVβ6 presented the highest affinity for PRGD2-based ligands. In situ, the expression of these integrins was significantly increased in OA compared to normal cartilage. Within osteophytes, the mean integrin expression score was significantly higher in blood vessels, fibrous areas, and cells from the bone lining than in osteocytes and cartilaginous zones. In vitro, the levels of integrin subunits were significantly increased during chondrocyte dedifferentiation (except for β6), fibrosis, and osteogenic differentiation as well as under inflammatory stimuli. In conclusion, anatomical zones (such as OA cartilage, intervertebral discs, and facet joint osteophytes) previously reported to show PRGD2 ligand uptake in vivo expressed increased levels of αVβ5, αVβ3, and β6 integrins, whose subunits are modulated in vitro by OA-associated conditions that increase fibrosis, inflammation, and osteogenic differentiation. These results suggest that the increased levels of integrins in OA compared to normal tissues favor PRGD2 uptake and might explain the molecular mechanism of OA imaging using the PRGD2-based ligand PET/CT.
Collapse
Affiliation(s)
- Edith Charlier
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | - Céline Deroyer
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | - Sophie Neuville
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | - Zelda Plener
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | - Olivier Malaise
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | - Federica Ciregia
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | | | - Gilles Reuter
- Department of Neurosurgery, CHULiège, Liège, Belgium
| | - Mallory Salvé
- Department of Nuclear Medicine, CHULiège, Liège, Belgium
| | - Nadia Withofs
- Department of Nuclear Medicine, CHULiège, Liège, Belgium
| | - Roland Hustinx
- Department of Nuclear Medicine, CHULiège, Liège, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| | - Michel G. Malaise
- Laboratory of Rheumatology, GIGA-I3, CHULiège, ULiège, Liège, Belgium
| |
Collapse
|
49
|
Chen Y, Li L, Zhang J. Cell migration inducing hyaluronidase 1 (CEMIP) activates STAT3 pathway to facilitate cell proliferation and migration in breast cancer. J Recept Signal Transduct Res 2020; 41:145-152. [PMID: 32757700 DOI: 10.1080/10799893.2020.1800732] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yu Chen
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| | - Lihong Li
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| | - Jinfan Zhang
- Department of Breast Surgery, The Affiliated Hospital of Putian University, Putian City, Fujian Province, China
| |
Collapse
|
50
|
Li C, Luo J, Xu X, Zhou Z, Ying S, Liao X, Wu K. Single cell sequencing revealed the underlying pathogenesis of the development of osteoarthritis. Gene 2020; 757:144939. [PMID: 32640306 DOI: 10.1016/j.gene.2020.144939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/25/2020] [Accepted: 07/01/2020] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is a chronic degenerative change with high incidence and leads to a lower quality of life and a larger socioeconomic burden. This study aimed to explore potential crucial genes and pathways associated with OA that can be used as potential biomarkers forearly treatment. Single-cell gene expression profile of 1464 chondrocytes and 192 fibroblasts in OA were downloaded from the public database (GSE104782 and GSE109449) for subsequent analysis. A total of eight clusters in chondrocytes and three clusters in fibroblasts of OA were identified using the Seurat pipeline and the "SingleR" package for cell-type annotation. Moreover, 44 common marker-genes between fibroblastic-like chondrocytes and fibroblasts were identified and the focal adhesions pathway was further identified as a significant potential mechanism of OA via functional enrichment analysis. Further, the reverse transcription quantitative real-time PCR (RT-qPCR) experiments at tissue's and cellular level confirmed that two key marker-genes (COL6A3 and ACTG1) might participate in the progression of OA. Summarily, we inferred that chondrocytes in OA might up-regulate the expression of COL6A3 and ACTG1 to complete fibroblasts transformation through the focal adhesion pathway. These findings are expected to gain a further insight into the development of OA fibrosis process and provide a promising target for treatment for early OA.
Collapse
Affiliation(s)
- Chenlu Li
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Luo
- Rheumatology Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Xu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zihao Zhou
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Senhong Ying
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xin Liao
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, China
| | - Keke Wu
- Wenzhou Center for Disease Control and Prevention, Wenzhou, China.
| |
Collapse
|