1
|
Bando Y, Bandow K, Sakiyama K, Nagasaka A, Suzuki K, Toda-Fujii M, Owada Y, Amano O. ETS1 promotes the expression of Ctsb and Mmp13 during the differentiation of septoclasts from pericytes. Cell Tissue Res 2025:10.1007/s00441-025-03979-x. [PMID: 40387924 DOI: 10.1007/s00441-025-03979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Septoclasts (SCs), which express both fatty acid-binding protein 5 and platelet-derived growth factor beta, are mononuclear cartilage-resorbing cells predominantly located at the chondro-osseous junction of the growth plate (GP). These cells originate from pericytes (PCs). Cathepsin B (CTSB) and matrix metalloproteinase-13 (MMP13), expressed in SCs, participate in the degradation of collagen and other cartilage matrices. This study aimed to investigate the involvement of the ETS proto-oncogene 1 (ETS1) in the transcription of Ctsb and Mmp13 during the differentiation of SCs from PCs. ETS1 was localized in SCs and a small number of PCs during development and postnatal stages. Upregulation of Ets1, Mmp13, Ctsb, and the Ets1-related genes, specificity protein 1 (Sp-1), jun proto-oncogene (c-Jun), and cAMP response element-binding protein-binding protein (Crebbp) in SCs compared with those in PCs was shown by RNA-seq analysis of samples isolated from the tibiae of 3-week-old postnatal mice. The Ets1-related proteins were localized ubiquitously in SCs and PCs in the GP. In primary SC cultures, the expression levels of Ctsb and Mmp13 were significantly reduced following treatment with Ets1 siRNA. Thus, our results revealed that ETS1 promoted the expression of Ctsb and Mmp13 in SCs during the differentiation of SCs from PCs.
Collapse
Affiliation(s)
- Yasuhiko Bando
- Division of Histology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan.
| | - Kenjiro Bandow
- Division of Biochemistry, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
| | - Koji Sakiyama
- Division of Anatomy, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
| | - Arata Nagasaka
- Division of Histology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
| | - Kaito Suzuki
- Division of Histology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
- Division of Oral and Maxillofacial Surgery, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
| | - Miyuki Toda-Fujii
- Division of Histology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 9808575, Japan
| | - Osamu Amano
- Division of Histology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 3500283, Japan
| |
Collapse
|
2
|
Behn A, Brendle S, Ehrnsperger M, Zborilova M, Grupp TM, Grifka J, Schäfer N, Grässel S. Filtered and unfiltered lipoaspirates reveal novel molecular insights and therapeutic potential for osteoarthritis treatment: a preclinical in vitro study. Front Cell Dev Biol 2025; 13:1534281. [PMID: 40083666 PMCID: PMC11903472 DOI: 10.3389/fcell.2025.1534281] [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: 11/25/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction Orthobiologics, such as autologous nanofat, are emerging as a potential treatment option for osteoarthritis (OA), a common degenerative joint causing pain and disability in the elderly. Nanofat, a minimally processed human fat graft rich in stromal vascular fraction (SVF) secretory factors, has shown promise in relieving pain. This study aimed to elucidate the molecular mechanisms underlying nanofat treatment of OA-affected cells and compare two filtration systems used for nanofat preparation. Methods Chondrocytes and synoviocytes were isolated from articular cartilage and synovium of 22 OA-patients. Lipoaspirates from 13 OA-patients were emulsified using the Adinizer® or Lipocube™ Nano filter systems to generate nanofat. The fluid phase of SVF from both filtered and unfiltered lipoaspirates was applied to OA-affected cells. Luminex multiplex ELISA were performed with lipoaspirates and cell supernatants alongside functional assays evaluating cell migration, proliferation, metabolic activity, and senescence. Results A total of 62 cytokines, chemokines, growth factors, neuropeptides, matrix-degrading enzymes, and complement components were identified in lipoaspirates. Among these, significant concentration differences were observed for TIMP-2, TGF-ß3, and complement component C3 between the filtered and unfiltered samples. Nanofat enhanced chondrocyte proliferation and migration, as well as synoviocyte migration and metabolic activity, while reducing chondrocyte metabolic activity. Pain-related factors like β-NGF, MCP-1, Substance P, VEGF, and αCGRP were reduced, while anti-inflammatory TGF-β1+3 increased and pro-inflammatory cytokines (IL-5, IL-7, IL-15, and IFN-γ) decreased. Nanofat also elevated secretion of complement components and TIMPs in both cell types. Notably, our results revealed no significant differences in cellular effects between sSVF filtered using the Adinizer® and Lipocube™ Nano systems, as well as compared to unfiltered sSVF. Discussion Here, we provide first insights into how autologous nanofat therapy may ameliorate OA by enhancing chondrocyte proliferation and synoviocyte migration while modulating inflammatory and pain-related factors. However, further research is needed to determine its effects on cartilage regeneration.
Collapse
Affiliation(s)
- Alissa Behn
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| | - Saskia Brendle
- Research and Development, Aesculap AG, Tuttlingen, Germany
- Department of Orthopaedic and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU Munich, Munich, Germany
| | - Marianne Ehrnsperger
- Department of Orthopedic Surgery, University of Regensburg, Asklepios, Germany
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Magdalena Zborilova
- Department of Orthopedic Surgery, University of Regensburg, Asklepios, Germany
| | - Thomas M. Grupp
- Research and Development, Aesculap AG, Tuttlingen, Germany
- Department of Orthopaedic and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU Munich, Munich, Germany
| | - Joachim Grifka
- Department of Orthopedic Surgery, University of Regensburg, Asklepios, Germany
- Department of Orthopedics and Ergonomics, Ostbayerische Technische Hochschule (OTH), Regensburg, Germany
| | - Nicole Schäfer
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| | - Susanne Grässel
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
- Department of Orthopedic Surgery, University of Regensburg, Asklepios, Germany
| |
Collapse
|
3
|
Dong DL, Jin GZ. Targeting Chondrocyte Hypertrophy as Strategies for the Treatment of Osteoarthritis. Bioengineering (Basel) 2025; 12:77. [PMID: 39851351 PMCID: PMC11760869 DOI: 10.3390/bioengineering12010077] [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: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by pain and functional impairment, which severely impacts the quality of life of middle-aged and elderly individuals. During normal bone development, chondrocyte hypertrophy is a natural physiological process. However, in the progression of OA, chondrocyte hypertrophy becomes one of its key pathological features. Although there is no definitive evidence to date confirming that chondrocyte hypertrophy is the direct cause of OA, substantial experimental data indicate that it plays an important role in the disease's pathogenesis. In this review, we first explore the mechanisms underlying chondrocyte hypertrophy in OA and offer new insights. We then propose strategies for inhibiting chondrocyte hypertrophy from the perspectives of targeting signaling pathways and tissue engineering, ultimately envisioning the future prospects of OA treatment.
Collapse
Affiliation(s)
- Da-Long Dong
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
| |
Collapse
|
4
|
Zeng D, Umar M, Zhu Z, Pan H, Lu WW, Xiao G, Chen Y, Tong L, Chen D. Development of novel osteoarthritis therapy by targeting AMPK-β-catenin-Runx2 signaling. Genes Dis 2025; 12:101247. [PMID: 39552787 PMCID: PMC11566674 DOI: 10.1016/j.gendis.2024.101247] [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: 09/25/2023] [Revised: 01/06/2024] [Accepted: 01/25/2024] [Indexed: 11/19/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating chronic joint disease affecting large populations of patients, especially the elderly. The pathological mechanisms of OA are currently unknown. Multiple risk factors are involved in OA development. Among these risk factors, alterations of mechanical loading in the joint leading to changes in biological signaling pathways have been known as a key event in OA development. The importance of AMPK-β-catenin-Runx2 signaling in the initiation and progression of OA has been recognized in recent years. In this review, we discuss the recent progress in understanding the role of this signaling pathway and the underlying interaction mechanisms during OA development. We also discuss the drug development aiming to target this signaling pathway for OA treatment.
Collapse
Affiliation(s)
- Daofu Zeng
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Muhammad Umar
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Zhenglin Zhu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Haobo Pan
- Shenzhen Healthemes Biotechnology Co., Ltd., Shenzhen, Guangdong 518071, China
| | - William W. Lu
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yan Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
5
|
Fan X, Ong LJY, Sun AR, Prasadam I. From polarity to pathology: Decoding the role of cell orientation in osteoarthritis. J Orthop Translat 2024; 49:62-73. [PMID: 39430130 PMCID: PMC11488446 DOI: 10.1016/j.jot.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 10/22/2024] Open
Abstract
UNLABELLED Cell polarity refers to the orientation of tissue and organelles within a cell and the direction of its function. It is one of the most critical characteristics of metazoans. The development, growth, and functional tissue distribution are closely related to holistic tissue or organ homeostasis. However, the connection between cell polarity and osteoarthritis (OA) is less well-known. In OA, multiple chondrocyte clusters and tissue disorganisation can be observed in the degraded cartilage tissue. The excessive upregulation of the planar cell polarity (PCP) signalling pathway leads to the loss of cell polarity and organisation in OA progression and aetiology. Recent research has become increasingly aware of the importance of cell polarity and its correlation with OA. Several cell polarity-related treatments have shed light on OA. A thorough understanding of cell polarity and OA would provide more insights for future investigations to treat this worldwide disease. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Understanding cell polarity, associated signalling pathways, organelle changes, and cell movement in the development of OA could lead to advances in precision medicine and enhanced treatment strategies for OA patients.
Collapse
Affiliation(s)
- Xiwei Fan
- Department of Orthopaedic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Louis Jun Ye Ong
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
| | - Antonia RuJia Sun
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Indira Prasadam
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
6
|
Sawauchi K, Fukui T, Oe K, Oda T, Yoshikawa R, Takase K, Inoue S, Nishida R, Kuroda R, Niikura T. Transcutaneous CO 2 application combined with low-intensity pulsed ultrasound accelerates bone fracture healing in rats. BMC Musculoskelet Disord 2024; 25:863. [PMID: 39472824 PMCID: PMC11523825 DOI: 10.1186/s12891-024-07976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Low-intensity pulsed ultrasound (LIPUS) is a non-invasive therapy that accelerates fracture healing. As a new treatment method for fracture, we recently reported that the transcutaneous application of CO2 accelerated fracture healing in association with promoting angiogenesis, blood flow, and endochondral ossification. We hypothesized that transcutaneous CO2 application, combined with LIPUS, would promote bone fracture healing more than the single treatment with either of them. METHODS Femoral shaft fractures were produced in 12-week-old rats. Animals were randomly divided into four groups: the combination of CO2 and LIPUS, CO2, LIPUS, and control groups. As the transcutaneous CO2 application, the limb was sealed in a CO2-filled bag after applying hydrogel that promotes CO2 absorption. Transcutaneous CO2 application and LIPUS irradiation were performed for 20 min/day, 5 days/week. At weeks 1, 2, 3, and 4 after the fractures, we assessed the fracture healing process using radiography, histology, immunohistochemistry, real-time PCR, and biomechanical assessment. RESULTS The fracture healing score using radiographs in the combination group was significantly higher than that in the control group at all time points and those in both the LIPUS and CO2 groups at weeks 1, 2, and 4. The degree of bone fracture healing in the histological assessment was significantly higher in the combination group than that in the control group at weeks 2, 3, and 4. In the immunohistochemical assessment, the vascular densities of CD31- and endomucin-positive microvessels in the combination group were significantly higher than those in the control and LIPUS groups at week 2. In the gene expression assessment, significant upregulation of runt-related transcription factor 2 (Runx2) and vascular endothelial growth factor (VEGF) was detected in the combination group compared to the LIPUS and CO2 monotherapy groups. In the biomechanical assessment, the ultimate stress was significantly higher in the combination group than in the LIPUS and CO2 groups. CONCLUSION The combination therapy of transcutaneous CO2 application and LIPUS had a superior effect in promoting fracture healing through the promotion of angiogenesis and osteoblast differentiation compared to monotherapy.
Collapse
Affiliation(s)
- Kenichi Sawauchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoaki Fukui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Oda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryo Yoshikawa
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kyohei Takase
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shota Inoue
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Ryota Nishida
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
- Department of Orthopaedic Surgery, Hyogo Prefectural Nishinomiya Hospital, Nishinomiya, Japan.
| |
Collapse
|
7
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [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: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
8
|
Roy HS, Murugesan P, Kulkarni C, Arora M, Nagar GK, Guha R, Chattopadhyay N, Ghosh D. On-demand release of a selective MMP-13 blocker from an enzyme-responsive injectable hydrogel protects cartilage from degenerative progression in osteoarthritis. J Mater Chem B 2024; 12:5325-5338. [PMID: 38669084 DOI: 10.1039/d3tb02871b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
In osteoarthritis (OA), the degradation of cartilage is primarily driven by matrix metalloprotease-13 (MMP-13). Hence, the inhibition of MMP-13 has emerged as an attractive target for OA treatment. Among the various approaches that are being explored for MMP-13 regulation, blocking of the enzyme with specific binding molecules appears to be a more promising strategy for preventing cartilage degeneration. To enhance effectiveness and ensure patient compliance, it is preferable for the binding molecule to exhibit sustained activity when administered directly into the joint. Herein, we present an enzyme-responsive hydrogel that was designed to exhibit on-demand, the sustained release of BI-4394, a potent and highly selective MMP-13 blocker. The stable and compatible hydrogel was prepared using triglycerol monostearate. The efficacy of the hydrogel to prevent cartilage damage was assessed in a rat model of OA induced by anterior cruciate ligament transection (ACLT). The results revealed that in comparison to the rats administrated weekly with intra-articular BI-4394, the hydrogel implanted rats had reduced levels of inflammation and bone erosion. In comparison to untreated control, the cartilage in animals administered with BI-4394/hydrogel exhibited significant levels of collagen-2 and aggrecan along with reduced MMP-13. Overall, this study confirmed the potential of BI-4394 delivery using an enzyme-responsive hydrogel as a promising treatment option to treat the early stages of OA by preventing further cartilage degradation.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Preethi Murugesan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chirag Kulkarni
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Malika Arora
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Geet Kumar Nagar
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Rajdeep Guha
- Division of Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in ASTHI, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
| | - Deepa Ghosh
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
9
|
Yao Y, Chen K, Pan Q, Gao H, Su W, Zheng S, Dong W, Qian D. Redifferentiation of genetically modified dedifferentiated chondrocytes in a microcavitary hydrogel. Biotechnol Lett 2024; 46:483-495. [PMID: 38523201 DOI: 10.1007/s10529-024-03475-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/11/2024] [Accepted: 02/15/2024] [Indexed: 03/26/2024]
Abstract
OBJECTIVES We genetically modified dedifferentiated chondrocytes (DCs) using lentiviral vectors and adenoviral vectors encoding TGF-β3 (referred to as transgenic groups below) and encapsulated these DCs in the microcavitary hydrogel and investigated the combinational effect on redifferentiation of the genetically manipulated DCs. RESULTS The Cell Counting Kit-8 data indicated that both transgenic groups exhibited significantly higher cell viability in the first week but inferior cell viability in the subsequent timepoints compared with those of the control group. Real-time polymerase chain reaction and western blot analysis results demonstrated that both transgenic groups had a better effect on redifferentiation to some extent, as evidenced by higher expression levels of chondrogenic genes, suggesting the validity of combination with transgenic DCs and the microcavitary hydrogel on redifferentiation. Although transgenic DCs with adenoviral vectors presented a superior extent of redifferentiation, they also expressed greater levels of the hypertrophic gene type X collagen. It is still worth further exploring how to deliver TGF-β3 more efficiently and optimizing the appropriate parameters, including concentration and duration. CONCLUSIONS The results demonstrated the better redifferentiation effect of DCs with the combinational use of transgenic TGF-β3 and a microcavitary alginate hydrogel and implied that DCs would be alternative seed cells for cartilage tissue engineering due to their easily achieved sufficient cell amounts through multiple passages and great potential to redifferentiate to produce cartilaginous extracellular matrix.
Collapse
Affiliation(s)
- Yongchang Yao
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
| | - Ke Chen
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Emergency Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
| | - Qian Pan
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Hui Gao
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Weixian Su
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- Guangdong Key Laboratory of Orthopaedic Technology and Implant Materials, Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Shicong Zheng
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Weiqiang Dong
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Dongyang Qian
- Department of Joint Surgery, The Key Laboratory of Advanced Interdisciplinary Studies Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| |
Collapse
|
10
|
Jain L, Bolam SM, Monk P, Munro JT, Tamatea J, Dalbeth N, Poulsen RC. Elevated glucose promotes MMP13 and ADAMTS5 production by osteoarthritic chondrocytes under oxygenated but not hypoxic conditions. J Cell Physiol 2024; 239:e31271. [PMID: 38595042 DOI: 10.1002/jcp.31271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024]
Abstract
Type 2 diabetes is linked with increased incidence and severity of osteoarthritis. The purpose of this study was to determine the effect of extracellular glucose within the normal blood glucose and hyperglycemic range on catabolic enzyme production by chondrocytes isolated from osteoarthritic (OA) and macroscopically normal (MN) human cartilage under oxygenated (18.9% oxygen) and hypoxic (1% oxygen) conditions. OA and MN chondrocytes were maintained in 4, 6, 8, or 10 mM glucose for 24 h. Glucose consumption, GLUT1 glucose transporter levels, MMP13 and ADAMTS5 production, and levels of RUNX2, a transcriptional regulator of MMP13, ADAMTS5, and GLUT1, were assessed by enzyme-linked assays, RT-qPCR and/or western blot. Under oxygenated conditions, glucose consumption and GLUT1 protein levels were higher in OA but not MN chondrocytes in 10 mM glucose compared to 4 mM. Both RNA and protein levels of MMP13 and ADAMTS5 were also higher in OA but not MN chondrocytes in 10 mM compared to 4 mM glucose under oxygenated conditions. Expression of RUNX2 was overall lower in MN than OA chondrocytes and there was no consistent effect of extracellular glucose concentration on RUNX2 levels in MN chondrocytes. However, protein (but not RNA) levels of RUNX2 were elevated in OA chondrocytes maintained in 10 mM versus 4 mM glucose under oxygenated conditions. In contrast, neither RUNX2 levels or MMP13 or ADAMTS5 expression were increased in OA chondrocytes maintained in 10 mM compared to 4 mM glucose in hypoxia. Elevated extracellular glucose leads to increased glucose consumption and increased RUNX2 protein levels, promoting production of MMP13 and ADAMTS5 by OA chondrocytes in oxygenated but not hypoxic conditions. These findings suggest that hyperglycaemia may exacerbate chondrocyte-mediated cartilage catabolism in the oxygenated superficial zone of cartilage in vivo in patients with undertreated type 2 diabetes, contributing to increased OA severity.
Collapse
Affiliation(s)
- Lekha Jain
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Scott M Bolam
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Paul Monk
- Department of Surgery, University of Auckland, Auckland, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Jacob T Munro
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Jade Tamatea
- Te Kupenga Hauora Māori, University of Auckland, Auckland, New Zealand
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and Tgfβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. eLife 2024; 13:e95640. [PMID: 38805545 PMCID: PMC11132684 DOI: 10.7554/elife.95640] [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: 01/02/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that CBFB (subunit of a heterodimeric Cbfβ/Runx1, Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfb in tamoxifen-induced Cbfbf/f;Col2a1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/Yap signaling and Tgfβ signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfb overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfb overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfb may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and Tgfβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfb overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
12
|
Lu L, Li J, Jiang X, Bai R. CXCR4/CXCL12 axis: "old" pathway as "novel" target for anti-inflammatory drug discovery. Med Res Rev 2024; 44:1189-1220. [PMID: 38178560 DOI: 10.1002/med.22011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/25/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024]
Abstract
Inflammation is the body's defense response to exogenous or endogenous stimuli, involving complex regulatory mechanisms. Discovering anti-inflammatory drugs with both effectiveness and long-term use safety is still the direction of researchers' efforts. The inflammatory pathway was initially identified to be involved in tumor metastasis and HIV infection. However, research in recent years has proved that the CXC chemokine receptor type 4 (CXCR4)/CXC motif chemokine ligand 12 (CXCL12) axis plays a critical role in the upstream of the inflammatory pathway due to its chemotaxis to inflammatory cells. Blocking the chemotaxis of inflammatory cells by CXCL12 at the inflammatory site may block and alleviate the inflammatory response. Therefore, developing CXCR4 antagonists has become a novel strategy for anti-inflammatory therapy. This review aimed to systematically summarize and analyze the mechanisms of action of the CXCR4/CXCL12 axis in more than 20 inflammatory diseases, highlighting its crucial role in inflammation. Additionally, the anti-inflammatory activities of CXCR4 antagonists were discussed. The findings might help generate new perspectives for developing anti-inflammatory drugs targeting the CXCR4/CXCL12 axis.
Collapse
Affiliation(s)
- Liuxin Lu
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junjie Li
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoying Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Saranya I, Akshaya R, Gomathi K, Mohanapriya R, He Z, Partridge N, Selvamurugan N. Circ_ST6GAL1-mediated competing endogenous RNA network regulates TGF-β1-stimulated matrix Metalloproteinase-13 expression via Runx2 acetylation in osteoblasts. Noncoding RNA Res 2024; 9:153-164. [PMID: 38035043 PMCID: PMC10686813 DOI: 10.1016/j.ncrna.2023.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
Transforming growth factor-beta1 (TGF-β1) stimulates matrix metalloproteinase-13 (MMP-13, a bone-remodeling gene) expression, and this effect requires p300-mediated Runx2 (Runt-related transcription factor 2) acetylation in osteoblasts. p300 and Runx2 are transcriptional coactivator and bone transcription factor, respectively, which play key roles in the regulation of bone-remodeling genes. Non-coding ribonucleic acids (ncRNAs), such as long ncRNAs (lncRNAs) and microRNAs (miRNAs), have been linked to both physiological and pathological bone states. In this study, we proposed that TGF-β1-mediated stimulation of MMP-13 expression is due to the downregulation of p300 targeting miRNAs in osteoblasts. We identified miR-130b-5p as one of the miRNAs downregulated by TGF-β1 in osteoblasts. Forced expression of miR-130b-5p decreased p300 expression, Runx2 acetylation, and MMP-13 expression in these cells. Furthermore, TGF-β1 upregulated circ_ST6GAL1, (a circular lncRNA) in osteoblasts; circRNA directly targeted miR-130b-5p. Antisense-mediated knockdown of circ_ST6GAL1 restored the function of miR-130b-5p, resulting in downregulation of p300, Runx2, and MMP-13 in these cells. Hence, our results suggest that TGF-β1 influences circ_ST6GAL1 to sponge and degrade miR-130b-5p, thereby promoting p300-mediated Runx2 acetylation for MMP-13 expression in osteoblasts. Thus, the circ_ST6GAL1/miR-130b-5p/p300 axis has potential significance in the treatment of bone and bone-related disorders.
Collapse
Affiliation(s)
- I. Saranya
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - R.L. Akshaya
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - K. Gomathi
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - R. Mohanapriya
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| | - Z. He
- Molecular Pathobiology, New York University College of Dentistry, New York, USA
| | - N.C. Partridge
- Molecular Pathobiology, New York University College of Dentistry, New York, USA
| | - N. Selvamurugan
- Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, India
| |
Collapse
|
14
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and TGFβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575763. [PMID: 38293189 PMCID: PMC10827176 DOI: 10.1101/2024.01.15.575763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that Cbfβ, (subunit of a heterodimeric Cbfβ/Runx1,Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfβ in tamoxifen-induced Cbfβf/fCol2α1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/YAP signaling and TGF-β signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfβ overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfβ overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfβ may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and TGFβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfβ overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
15
|
Ratanasereeprasert N, Hsu LF, Wang SK, Jane Yao CC. Orthodontically induced changes to the genetic profile in periodontal ligament tissue and cytokine release in gingival crevicular fluid - A pilot investigation. J Dent Sci 2024; 19:387-396. [PMID: 38303827 PMCID: PMC10829649 DOI: 10.1016/j.jds.2023.07.038] [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: 07/28/2023] [Revised: 07/31/2023] [Indexed: 02/03/2024] Open
Abstract
Background/purpose It has been known that genetic factors influence orthodontic tooth movement, however, scientific research on humans is lacking. Therefore, this study aimed to investigate dynamic changes to the genetic profile in human periodontal ligament (PDL) tissue and cytokine release in gingival crevicular fluid (GCF) during the first 28 days of orthodontic treatment. Materials and methods Fifteen teeth from three patients were recruited. Full-mouth fixed appliances with extraction of four premolars and one maxillary third molar was planned for orthodontic treatment. GCF collection and tooth extraction were performed following force application for 0, 1, 3, 7, and 28 days. GCF was analyzed using multiplex immunoassay for 27 cytokines. PDL tissue was collected after extraction and submitted for RNA exome-sequencing using Illumina sequencing platform. Further analysis of differentially expressed genes (DEGs), gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and heatmaps were conducted. Results GCF cytokine levels varied among three patients; some patients exhibited a peak cytokine level on Day 0 whereas others did so on Days 1-3. In RNA exome sequencing data, GO and KEGG analyses showed that genes associated with sensory receptors were upregulated on Day 1, genes involved in bone remodeling were upregulated on Days 3 and 28, and genes related to osteoclast differentiation were upregulated on Day 7. Conclusion RNA sequencing data demonstrate that the specific types of genes are expressed at different time points, whereas the data on cytokine changes show a large variation in concentration levels and dynamic change patterns among the patients.
Collapse
Affiliation(s)
| | - Li-Fang Hsu
- Department of Dentistry, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Shih-Kai Wang
- Department of Dentistry, School of Dentistry, National Taiwan University, Department of Pediatric Dentistry, National Taiwan University Children's Hospital, Taipei, Taiwan
| | - Chung-Chen Jane Yao
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Division of Orthodontics and Dentofacial Orthopedics, Dental Department, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
Hollander JM, Goraltchouk A, Liu J, Xu E, Luppino F, McAlindon TE, Zeng L, Seregin A. Single Injection AAV2-FGF18 Gene Therapy Reduces Cartilage Loss and Subchondral Bone Damage in a Mechanically Induced Model of Osteoarthritis. Curr Gene Ther 2024; 24:331-345. [PMID: 38783531 DOI: 10.2174/0115665232275532231213063634] [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: 08/04/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 05/25/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a highly debilitating, degenerative pathology of cartilaginous joints affecting over 500 million people worldwide. The global economic burden of OA is estimated at $260-519 billion and growing, driven by aging global population and increasing rates of obesity. To date, only the multi-injection chondroanabolic treatment regimen of Fibroblast Growth Factor 18 (FGF18) has demonstrated clinically meaningful disease-modifying efficacy in placebo-controlled human trials. Our work focuses on the development of a novel single injection disease-modifying gene therapy, based on FGF18's chondroanabolic activity. METHODS OA was induced in Sprague-Dawley rats using destabilization of the medial meniscus (DMM) (3 weeks), followed by intra-articular treatment with 3 dose levels of AAV2-FGF18, rh- FGF18 protein, and PBS. Durability, redosability, and biodistribution were measured by quantifying nLuc reporter bioluminescence. Transcriptomic analysis was performed by RNA-seq on cultured human chondrocytes and rat knee joints. Morphological analysis was performed on knee joints stained with Safranin O/Fast Green and anti-PRG antibody. RESULTS Dose-dependent reductions in cartilage defect size were observed in the AAV2-FGF18- treated joints relative to the vehicle control. Total defect width was reduced by up to 76% and cartilage thickness in the thinnest zone was increased by up to 106%. Morphologically, the vehicle- treated joints exhibited pronounced degeneration, ranging from severe cartilage erosion and bone void formation, to subchondral bone remodeling and near-complete subchondral bone collapse. In contrast, AAV2-FGF18-treated joints appeared more anatomically normal, with only regional glycosaminoglycan loss and marginal cartilage erosion. While effective at reducing cartilage lesions, treatment with rhFGF18 injections resulted in significant joint swelling (19% increase in diameter), as well as a decrease in PRG4 staining uniformity and intensity. In contrast to early-timepoint in vitro RNA-seq analysis, which showed a high degree of concordance between protein- and gene therapy-treated chondrocytes, in vivo transcriptomic analysis, revealed few gene expression changes following protein treatment. On the other hand, the gene therapy treatment exhibited a high degree of durability and localization over the study period, upregulating several chondroanabolic genes while downregulating OA- and fibrocartilage-associated markers. CONCLUSION FGF18 gene therapy treatment of OA joints can provide benefits to both cartilage and subchondral bone, with a high degree of localization and durability.
Collapse
Affiliation(s)
- Judith M Hollander
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| | - Alex Goraltchouk
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| | - Jingshu Liu
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
| | - Ellyn Xu
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
| | - Francesco Luppino
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| | - Timothy E McAlindon
- Division of Rheumatology, Immunology, and Allergy, Tufts Medical Center, Boston, MA, United States of America
| | - Li Zeng
- Department of Immunology, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA, United States of America
| | - Alexey Seregin
- Remedium Bio, Inc. 1116 Great Plain Ave, Suite 203, Needham, MA, United States of America
| |
Collapse
|
17
|
Larder CE, Iskandar MM, Kubow S. Collagen Hydrolysates: A Source of Bioactive Peptides Derived from Food Sources for the Treatment of Osteoarthritis. MEDICINES (BASEL, SWITZERLAND) 2023; 10:50. [PMID: 37755240 PMCID: PMC10538231 DOI: 10.3390/medicines10090050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 09/28/2023]
Abstract
Osteoarthritis (OA) is the most common joint disorder, with a social and financial burden that is expected to increase in the coming years. Currently, there are no effective medications to treat it. Due to limited treatment options, patients often resort to supplements, such as collagen hydrolysates (CHs). CHs are products with low molecular weight (MW) peptides, often between 3 and 6 kDa, and are a result of industrialized processed collagen. Collagen extraction is often a by-product of the meat industry, with the main source for collagen-based products being bovine, although it can also be obtained from porcine and piscine sources. CHs have demonstrated positive results in clinical trials related to joint health, such as decreased joint pain, increased mobility, and structural joint improvements. The bioactivity of CHs is primarily attributed to their bioactive peptide (BAP) content. However, there are significant knowledge gaps regarding the digestion, bioavailability, and bioactivity of CH-derived BAPs, and how different CH products compare in that regard. The present review discusses CHs and their BAP content as potential treatments for OA.
Collapse
Affiliation(s)
- Christina E. Larder
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC H9X 3V9, Canada; (C.E.L.); (M.M.I.)
- Corporation Genacol Canada Inc., Blainville, QC J7C 6B4, Canada
| | - Michèle M. Iskandar
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC H9X 3V9, Canada; (C.E.L.); (M.M.I.)
| | - Stan Kubow
- School of Human Nutrition, McGill University, Ste-Anne-de-Bellevue, QC H9X 3V9, Canada; (C.E.L.); (M.M.I.)
| |
Collapse
|
18
|
Poudel SB, Ruff RR, Yildirim G, Dixit M, Michot B, Gibbs JL, Ortiz SD, Kopchick JJ, Kirsch T, Yakar S. Excess Growth Hormone Triggers Inflammation-Associated Arthropathy, Subchondral Bone Loss, and Arthralgia. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:829-842. [PMID: 36870529 PMCID: PMC10284029 DOI: 10.1016/j.ajpath.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023]
Abstract
Growth hormone (GH) is a key mediator of skeletal growth. In humans, excess GH secretion due to pituitary adenoma, seen in patients with acromegaly, results in severe arthropathies. This study investigated the effects of long-term excess GH on the knee joint tissues. One year-old wild-type (WT) and bovine GH (bGH) transgenic mice were used as a model for excess GH. bGH mice showed increased sensitivity to mechanical and thermal stimuli, compared with WT mice. Micro-computed tomography analyses of the distal femur subchondral bone revealed significant reductions in trabecular thickness and significantly reduced bone mineral density of the tibial subchondral bone-plate associated with increased osteoclast activity in both male and female bGH compared with WT mice. bGH mice showed severe loss of matrix from the articular cartilage, osteophytosis, synovitis, and ectopic chondrogenesis. Articular cartilage loss in the bGH mice was associated with elevated markers of inflammation and chondrocyte hypertrophy. Finally, hyperplasia of synovial cells was associated with increased expression of Ki-67 and diminished p53 levels in the synovium of bGH mice. Unlike the low-grade inflammation seen in primary osteoarthritis, arthropathy caused by excess GH affects all joint tissues and triggers severe inflammatory response. Data from this study suggest that treatment of acromegalic arthropathy should involve inhibition of ectopic chondrogenesis and chondrocyte hypertrophy.
Collapse
Affiliation(s)
- Sher B Poudel
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Ryan R Ruff
- Department of Epidemiology and Health Promotion, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Gozde Yildirim
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Manisha Dixit
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York
| | - Benoit Michot
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Jennifer L Gibbs
- Department of Restorative Dentistry and Biomaterials Sciences, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Silvana D Ortiz
- Department of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - John J Kopchick
- Department of Biomedical Sciences, Edison Biotechnology Institute, Ohio University, Athens, Ohio
| | - Thorsten Kirsch
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, New York; Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, New York
| | - Shoshana Yakar
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, New York.
| |
Collapse
|
19
|
Qu M, Chen M, Gong W, Huo S, Yan Q, Yao Q, Lai Y, Chen D, Wu X, Xiao G. Pip5k1c Loss in Chondrocytes Causes Spontaneous Osteoarthritic Lesions in Aged Mice. Aging Dis 2023; 14:502-514. [PMID: 37008048 PMCID: PMC10017150 DOI: 10.14336/ad.2022.0828] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease affecting the older populations globally. Phosphatidylinositol-4-phosphate 5-kinase type-1 gamma (Pip5k1c), a lipid kinase catalyzing the synthesis of phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2), is involved in various cellular processes, such as focal adhesion (FA) formation, cell migration, and cellular signal transduction. However, whether Pip5k1c plays a role in the pathogenesis of OA remains unclear. Here we show that inducible deletion of Pip5k1c in aggrecan-expressing chondrocytes (cKO) causes multiple spontaneous OA-like lesions, including cartilage degradation, surface fissures, subchondral sclerosis, meniscus deformation, synovial hyperplasia, and osteophyte formation in aged (15-month-old) mice, but not in adult (7-month-old) mice. Pip5k1c loss promotes extracellular matrix (ECM) degradation, chondrocyte hypertrophy and apoptosis, and inhibits chondrocyte proliferation in the articular cartilage of aged mice. Pip5k1c loss dramatically downregulates the expressions of several key FA proteins, including activated integrin β1, talin, and vinculin, and thus impairs the chondrocyte adhesion and spreading on ECM. Collectively, these findings suggest that Pip5k1c expression in chondrocytes plays a critical role in maintaining articular cartilage homeostasis and protecting against age-related OA.
Collapse
Affiliation(s)
- Minghao Qu
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Shaochuan Huo
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Qinnan Yan
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Yumei Lai
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
20
|
Cai Y, Wang Z, Liao B, Sun Z, Zhu P. Anti-inflammatory and Chondroprotective Effects of Platelet-derived Growth Factor-BB on Osteoarthritis Rat Models. J Gerontol A Biol Sci Med Sci 2023; 78:51-59. [PMID: 35640164 DOI: 10.1093/gerona/glac118] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Indexed: 01/31/2023] Open
Abstract
Osteoarthritis (OA) is a common and challenging joint disease that mainly affects the diarthrodial joints. Traditionally, except for surgery for severe cases, treatments for OA mainly focus on relieving pain and improving joint function. However, these treatments are not effective for cartilage repair and induce only symptomatic relief. Platelet-derived growth factor (PDGF)-BB, a member of the PDGF cytokine family, has been proved to have effects on protecting the chondrocytes via multiple mechanisms. In this study, we further focused on the effects of PDGF-BB on OA and found that PDGF-BB could attenuate OA development by inhibiting inflammation and enhancing cell proliferation via JAK2/STAT3, PI3K/AKT, and p38 signaling pathways and PKA-mediated regulation of SOX-9/RunX-2. This article demonstrates the feasibility of PDGF-BB application as a treatment for OA. This is the first article that reports that PDGF-BB attenuates OA development via PKA-mediated regulation of SOX-9 and RunX-2.
Collapse
Affiliation(s)
- Yu Cai
- Department of Rehabilitation, Wuhan Fourth Hospital, Wuhan, China
| | - Zhengchao Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bokai Liao
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, China
| | - Zhenxing Sun
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengfei Zhu
- Department of Cardiology, Wuhan Fourth Hospital, Wuhan, China
| |
Collapse
|
21
|
Du X, Cai L, Xie J, Zhou X. The role of TGF-beta3 in cartilage development and osteoarthritis. Bone Res 2023; 11:2. [PMID: 36588106 PMCID: PMC9806111 DOI: 10.1038/s41413-022-00239-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/25/2022] [Accepted: 11/03/2022] [Indexed: 01/03/2023] Open
Abstract
Articular cartilage serves as a low-friction, load-bearing tissue without the support with blood vessels, lymphatics and nerves, making its repair a big challenge. Transforming growth factor-beta 3 (TGF-β3), a vital member of the highly conserved TGF-β superfamily, plays a versatile role in cartilage physiology and pathology. TGF-β3 influences the whole life cycle of chondrocytes and mediates a series of cellular responses, including cell survival, proliferation, migration, and differentiation. Since TGF-β3 is involved in maintaining the balance between chondrogenic differentiation and chondrocyte hypertrophy, its regulatory role is especially important to cartilage development. Increased TGF-β3 plays a dual role: in healthy tissues, it can facilitate chondrocyte viability, but in osteoarthritic chondrocytes, it can accelerate the progression of disease. Recently, TGF-β3 has been recognized as a potential therapeutic target for osteoarthritis (OA) owing to its protective effect, which it confers by enhancing the recruitment of autologous mesenchymal stem cells (MSCs) to damaged cartilage. However, the biological mechanism of TGF-β3 action in cartilage development and OA is not well understood. In this review, we systematically summarize recent progress in the research on TGF-β3 in cartilage physiology and pathology, providing up-to-date strategies for cartilage repair and preventive treatment.
Collapse
Affiliation(s)
- Xinmei Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
22
|
Cai Y, Wu C, Ou Q, Zeng M, Xue S, Chen J, Lu Y, Ding C. Enhanced osteoarthritis therapy by nanoengineered mesenchymal stem cells using biomimetic CuS nanoparticles loaded with plasmid DNA encoding TGF-β1. Bioact Mater 2023; 19:444-457. [PMID: 35574050 PMCID: PMC9079106 DOI: 10.1016/j.bioactmat.2022.04.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/11/2022] [Accepted: 04/21/2022] [Indexed: 11/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) therapy shows the potential benefits to relieve clinical symptoms of osteoarthritis (OA), but it is uncertain if it can repair articular cartilage lesions — the main pathology of OA. Here, we prepared biomimetic cupper sulfide@phosphatidylcholine (CuS@PC) nanoparticles (NPs) loaded with plasmid DNA (pDNA) encoding transforming growth factor-beta 1 (TGF-β1) to engineer MSCs for enhanced OA therapy via cartilage regeneration. We found that the NPs not only promoted cell proliferation and migration, but also presented a higher pDNA transfection efficiency relative to commercial transfection reagent lipofectamine 3000. The resultant CuS/TGF-β1@PC NP-engineered MSCs (termed CTP-MSCs) were better than pure MSCs in terms of chondrogenic gene expression, glycosaminoglycan deposition and type II collagen formation, favoring cartilage repair. Further, CTP-MSCs inhibited extracellular matrix degradation in interleukin-1β-induced chondrocytes. Consequently, intraarticular administration of CTP-MSCs significantly enhanced the repair of damaged cartilage, whereas pure MSCs exhibited very limited effects on cartilage regeneration in destabilization of the medial meniscus (DMM) surgical instability mice. Hence, this work provides a new strategy to overcome the limitation of current stem cell therapy in OA treatment through developing more effective nanoengineered MSCs. Biomimetic CuS nanoparticles (NPs) loaded with TGF-β1 pDNA are prepared for nanoengineering of MSCs. CuS/TGF-β1@PC NPs are more efficient than commercial transfection agent in terms of pDNA transfection. The NP-engineered CTP-MSCs exhibit enhanced migration, chondrogenesis and inhibition of ECM degradation. CTP-MSCs effectively treat osteoarthritis (OA) mice models via cartilage regeneration.
Collapse
|
23
|
Li C, Li W, Pu G, Wu J, Qin F. Exosomes derived from miR-338-3p-modified adipose stem cells inhibited inflammation injury of chondrocytes via targeting RUNX2 in osteoarthritis. J Orthop Surg Res 2022; 17:567. [PMID: 36572886 PMCID: PMC9791748 DOI: 10.1186/s13018-022-03437-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease that is one of the main causes of disability in middle-aged and elderly people. Adipose stem cell (ASC)-derived exosomes (ASC-Exo) could repair cartilage damage and treat OA. MiRNA-338-3p expression was confirmed to play a role in inhibiting proinflammatory cytokines. Herein, we aimed to explore the mechanism by which exosomes derived from miR-338-3p overexpressing ASCs protects chondrocytes from interleukin (IL)-1β-induced chondrocyte change. METHODS Exosomes were extracted from ASCs transfected with miR-338-3p or its antisense inhibitor. The ASC-Exos (miR-338-3p silencing/overexpression) were incubated with IL-1β-induced ATDC5 cells, followed by evaluation of the chondrocyte proliferation, degradation, and inflammation injury. RESULTS In vitro results revealed that ASC-Exos inhibited the expression of prostaglandin E2 (PGE2), IL-6, IL-1β, and TNF-α, as well as promoted the proliferation of ATDC5 cells. Moreover, ASC-Exos inhibited inflammation injury and degradation of ATDC5 cells by transferring miR-338-3p. Luciferase reporter assays showed that RUNX2 was a target gene of miR-338-3p. Additionally, RUNX2 overexpression in ATDC5 cells reversed the protective effect of miR-338-3p on chondrocytes. Taken together, this study demonstrated that exosomes secreted from miR-338-3p-modified ASCs were effective in the repair of IL-1β-induced chondrocyte change by inhibiting RUNX2 expression. CONCLUSIONS Our result provided valuable data for understanding the mechanism of ASC-Exos in OA treatment.
Collapse
Affiliation(s)
- ChunLiang Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Wei Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - GengZang Pu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - JingWen Wu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Feng Qin
- grid.459333.bDepartment of Endocrinology, Qinghai University Affiliated Hospital, Chengxi District, No. 6, Xichuan South Road, Xining, 810006 Qinghai China
| |
Collapse
|
24
|
Meera M. Recent advances in the pharmacotherapy of osteoarthritis. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.84951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: Osteoarthritis (OA) is a common debilitating disease affecting the geriatric population. Management of osteoarthritis is a challenge for orthopedicians because till date there has been no such drug that can completely cure the disease or at least retard/arrest the disease progression. In addition to the currently available treatment options for OA like NSAIDs, opioids, nutraceuticals (glucosamine sulphate and chondroitin sulphate), many new drugs are being discovered or repurposed for use in osteoarthritis. Most of these recent drugs aim at retarding the disease progression rather than providing just a symptomatic relief.
Materials and methods: All relevant articles regarding approved new drugs and pipeline drugs for osteoarthritis published between 2012–2021 were analysed. Those included animal studies as well as clinical trials. Some older articles were also referred to, provided they highlighted any significant data. The obtained data were analysed and compiled.
Results and discussion: Broadly the recent drugs for OA can be classified based upon their site of action as (i) drugs targeting articular cartilage, (ii) drugs targeting inflammation, (iii) drugs targeting the subchondral bone, and (iv) drugs for relieving pain. Ranging from in vitro studies to clinical trials, these drugs are in various phases of drug discovery. Early diagnosis of OA and its management with a drug that retards disease progression rather than prescribing just a symptom reliever is very much necessary in the current situation.
Conclusion: Need for new drugs for OA is increasing day by day. More number of clinical trials with larger sample sizes alone can satisfy the need of disease modifying drugs for OA. This review provides a deep insight into all the recent advances in the pharmacotherapy of osteoarthritis.
Graphical abstract:
Collapse
|
25
|
Shi Y, Shao J, Zhang Z, Zhang J, Lu H. Effect of condylar chondrocyte exosomes on condylar cartilage osteogenesis in rats under tensile stress. Front Bioeng Biotechnol 2022; 10:1061855. [PMID: 36561044 PMCID: PMC9766957 DOI: 10.3389/fbioe.2022.1061855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Functional orthoses are commonly used to treat skeletal Class II malocclusion, but the specific mechanism through which they do this has been a challenging topic in orthodontics. In the present study, we aimed to explore the effect of tensile stress on the osteogenic differentiation of condylar chondrocytes from an exosomal perspective. Methods: We cultured rat condylar chondrocytes under resting and tensile stress conditions and subsequently extracted cellular exosomes from them. We then screened miRNAs that were differentially expressed between the two exosome extracts by high-throughput sequencing and performed bioinformatics analysis and osteogenesis-related target gene prediction using the TargetScan and miRanda softwares. Exosomes cultured under resting and tensile stress conditions were co-cultured with condylar chondrocytes for 24 h to form the Control-Exo and Force-Exo exosome groups, respectively. Quantitative real time PCR(RT-qPCR) and western blotting were then used to determine the mRNA and protein expression levels of Runx2 and Sox9 in condylar chondrocytes. Results: The mRNA and protein expression levels of Runx2 and Sox9 in the Force-Exo group were significantly higher than those in the Control-Exo group (p < 0.05). The differential miRNA expression results were consistent with our sequencing results. Bioinformatics analysis and target gene prediction results showed that the main biological processes and molecular functions involved in differential miRNA expression in exosomes under tensile stress were biological processes and protein binding, respectively. Kyoto Gene and Genome Data Bank (KEGG) pathway enrichment analysis showed significant enrichment of differentially expressed miRNAs in the mTOR signaling pathway. The differentially expressed miRNAs were found to target osteogenesis-related genes. Conclusion: These results suggest that stimulation of rat condylar chondrocytes with tensile stress can alter the expression levels of certain miRNAs in their exosomes and promote their osteogenic differentiation. Exosomes under tensile stress culture conditions thus have potential applications in the treatment of Osteoarthritis (OA).
Collapse
Affiliation(s)
- Yuan Shi
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Shao
- Department of Stomatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Zanzan Zhang
- Department of Stomatology, Ningbo No. 2 Hospital, Ningbo, China
| | - Jianan Zhang
- Department of Dentistry, Center of Orthodontics, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Haiping Lu
- Department of Stomatology, Zhejiang Chinese Medical University, Hangzhou, China,*Correspondence: Haiping Lu,
| |
Collapse
|
26
|
Li J, Jiang H, Lv Z, Sun Z, Cheng C, Tan G, Wang M, Liu A, Sun H, Guo H, Chen F, Liu Z, Fei Y, Liu Y, Wu R, Xu X, Yan W, Jiang Q, Shi D. Articular fibrocartilage-targeted therapy by microtubule stabilization. SCIENCE ADVANCES 2022; 8:eabn8420. [PMID: 36399569 PMCID: PMC9674280 DOI: 10.1126/sciadv.abn8420] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
The fibrocartilage presented on the joint surface was caused by cartilage injury or degeneration. There is still a lack of effective strategies for fibrocartilage. Here, we hypothesized that the fibrocartilage could be viewed as a raw material for the renewal of hyaline cartilage and proposed a previously unidentified strategy of cartilage regeneration, namely, "fibrocartilage hyalinization." Cytoskeleton remodeling plays a vital role in modifying the cellular phenotype. We identified that microtubule stabilization by docetaxel repressed cartilage fibrosis and increased the hyaline cartilage extracellular matrix. We further designed a fibrocartilage-targeted negatively charged thermosensitive hydrogel for the sustained delivery of docetaxel, which promoted fibrocartilage hyalinization in the cartilage defect model. Moreover, the mechanism of fibrocartilage hyalinization by microtubule stabilization was verified as the inhibition of Sparc (secreted protein acidic and rich in cysteine). Together, our study suggested that articular fibrocartilage-targeted therapy in situ was a promising strategy for hyaline cartilage repair.
Collapse
Affiliation(s)
- Jiawei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Huiming Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu, P.R. China
| | - Zhongyang Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Ziying Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Chaoqun Cheng
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, 210023 Jiangsu, P.R. China
| | - Guihua Tan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Maochun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Anlong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Heng Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Hu Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Fufei Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Zizheng Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Yuxiang Fei
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Yuan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Rui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Xingquan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Wenjin Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| | - Dongquan Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School Nanjing, Nanjing, 210008 Jiangsu, P.R. China
| |
Collapse
|
27
|
Nagata K, Hojo H, Chang SH, Okada H, Yano F, Chijimatsu R, Omata Y, Mori D, Makii Y, Kawata M, Kaneko T, Iwanaga Y, Nakamoto H, Maenohara Y, Tachibana N, Ishikura H, Higuchi J, Taniguchi Y, Ohba S, Chung UI, Tanaka S, Saito T. Runx2 and Runx3 differentially regulate articular chondrocytes during surgically induced osteoarthritis development. Nat Commun 2022; 13:6187. [PMID: 36261443 PMCID: PMC9581901 DOI: 10.1038/s41467-022-33744-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
The Runt-related transcription factor (Runx) family plays various roles in the homeostasis of cartilage. Here, we examined the role of Runx2 and Runx3 for osteoarthritis development in vivo and in vitro. Runx3-knockout mice exhibited accelerated osteoarthritis following surgical induction, accompanied by decreased expression of lubricin and aggrecan. Meanwhile, Runx2 conditional knockout mice showed biphasic phenotypes: heterozygous knockout inhibited osteoarthritis and decreased matrix metallopeptidase 13 (Mmp13) expression, while homozygous knockout of Runx2 accelerated osteoarthritis and reduced type II collagen (Col2a1) expression. Comprehensive transcriptional analyses revealed lubricin and aggrecan as transcriptional target genes of Runx3, and indicated that Runx2 sustained Col2a1 expression through an intron 6 enhancer when Sox9 was decreased. Intra-articular administration of Runx3 adenovirus ameliorated development of surgically induced osteoarthritis. Runx3 protects adult articular cartilage through extracellular matrix protein production under normal conditions, while Runx2 exerts both catabolic and anabolic effects under the inflammatory condition.
Collapse
Affiliation(s)
- Kosei Nagata
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Hironori Hojo
- grid.26999.3d0000 0001 2151 536XCenter for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Song Ho Chang
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Hiroyuki Okada
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan ,grid.26999.3d0000 0001 2151 536XCenter for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Fumiko Yano
- grid.26999.3d0000 0001 2151 536XBone and Cartilage Regenerative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Ryota Chijimatsu
- grid.26999.3d0000 0001 2151 536XBone and Cartilage Regenerative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Yasunori Omata
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan ,grid.26999.3d0000 0001 2151 536XBone and Cartilage Regenerative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Daisuke Mori
- grid.26999.3d0000 0001 2151 536XBone and Cartilage Regenerative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Yuma Makii
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Manabu Kawata
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Taizo Kaneko
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Yasuhide Iwanaga
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Hideki Nakamoto
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Yuji Maenohara
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Naohiro Tachibana
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Hisatoshi Ishikura
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Junya Higuchi
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Yuki Taniguchi
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Shinsuke Ohba
- grid.26999.3d0000 0001 2151 536XCenter for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan ,grid.174567.60000 0000 8902 2273Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588 Japan
| | - Ung-il Chung
- grid.174567.60000 0000 8902 2273Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588 Japan
| | - Sakae Tanaka
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| | - Taku Saito
- grid.26999.3d0000 0001 2151 536XSensory & Motor System Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655 Japan
| |
Collapse
|
28
|
Downregulation of miR-30b-5p Facilitates Chondrocyte Hypertrophy and Apoptosis via Targeting Runx2 in Steroid-Induced Osteonecrosis of the Femoral Head. Int J Mol Sci 2022; 23:ijms231911275. [PMID: 36232582 PMCID: PMC9570061 DOI: 10.3390/ijms231911275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
As a widely used steroid hormone medicine, glucocorticoids have the potential to cause steroid-induced osteonecrosis of the femoral head (SONFH) due to mass or long-term use. The non-coding RNA hypothesis posits that they may contribute to the destruction and dysfunction of cartilages as a possible etiology of SONFH. MiR-30b-5p was identified as a regulatory factor in cartilage degeneration caused by methylprednisolone (MPS) exposure in our study through cell transfection. The luciferase reporter assay confirmed that miR-30b-5p was downregulated and runt-related transcription factor 2 (Runx2) was mediated by miR-30b-5p. The nobly increased expression of matrix metallopeptidase 13 (MMP13) and type X collagen (Col10a1) as Runx2 downstream genes contributed to the hypertrophic differentiation of chondrocytes, and the efficiently upregulated level of matrix metallopeptidase 9 (MMP9) may trigger chondrocyte apoptosis with MPS treatments. The cell transfection experiment revealed that miR-30b-5p inhibited chondrocyte hypertrophy and suppressed MPS-induced apoptosis. As a result, our findings showed that miR-30b-5p modulated Runx2, MMP9, MMP13, and Col10a1 expression, thereby mediating chondrocyte hypertrophic differentiation and apoptosis during the SONFH process. These findings revealed the mechanistic relationship between non-coding RNA and SONFH, providing a comprehensive understanding of SONFH and other bone diseases.
Collapse
|
29
|
Liu HW, Su WT, Liu CY, Huang CC. Highly Organized Porous Gelatin-Based Scaffold by Microfluidic 3D-Foaming Technology and Dynamic Culture for Cartilage Tissue Engineering. Int J Mol Sci 2022; 23:ijms23158449. [PMID: 35955581 PMCID: PMC9369316 DOI: 10.3390/ijms23158449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
A gelatin-based hydrogel scaffold with highly uniform pore size and biocompatibility was fabricated for cartilage tissue engineering using microfluidic 3D-foaming technology. Mainly, bubbles with different diameters, such as 100 μm and 160 μm, were produced by introducing an optimized nitrogen gas and gelatin solution at an optimized flow rate, and N2/gelatin bubbles were formed. Furthermore, a cross-linking agent (1-ethyl-3-(3-dimethyl aminopropyl)-carbodiimide, EDC) was employed for the cross-linking reaction of the gelatin-based hydrogel scaffold with uniform bubbles, and then the interface between the close cells were broken by degassing. The pore uniformity of the gelatin-based hydrogel scaffolds was confirmed by use of a bright field microscope, conjugate focus microscope and scanning electron microscope. The in vitro degradation rate, mechanical properties, and swelling rate of gelatin-based hydrogel scaffolds with highly uniform pore size were studied. Rabbit knee cartilage was cultured, and its extracellular matrix content was analyzed. Histological analysis and immunofluorescence staining were employed to confirm the activity of the rabbit knee chondrocytes. The chondrocytes were seeded into the resulting 3D porous gelatin-based hydrogel scaffolds. The growth conditions of the chondrocyte culture on the resulting 3D porous gelatin-based hydrogel scaffolds were evaluated by MTT analysis, live/dead cell activity analysis, and extracellular matrix content analysis. Additionally, a dynamic culture of cartilage tissue was performed, and the expression of cartilage-specific proteins within the culture time was studied by immunofluorescence staining analysis. The gelatin-based hydrogel scaffold encouraged chondrocyte proliferation, promoting the expression of collagen type II, aggrecan, and sox9 while retaining the structural stability and durability of the cartilage after dynamic compression and promoting cartilage repair.
Collapse
Affiliation(s)
- Hsia-Wei Liu
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (H.-W.L.); (C.-Y.L.)
- Graduate Institute of Applied Science and Engineering, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Wen-Ta Su
- Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 106344, Taiwan;
| | - Ching-Yi Liu
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (H.-W.L.); (C.-Y.L.)
| | - Ching-Cheng Huang
- Department of Biomedical Engineering, Ming-Chuan University, Taoyuan 333321, Taiwan
- PARSD Biomedical Material Research Center, Taichung 407428, Taiwan
- Correspondence:
| |
Collapse
|
30
|
Lai Y, Zheng W, Qu M, Xiao CC, Chen S, Yao Q, Gong W, Tao C, Yan Q, Zhang P, Wu X, Xiao G. Kindlin-2 loss in condylar chondrocytes causes spontaneous osteoarthritic lesions in the temporomandibular joint in mice. Int J Oral Sci 2022; 14:33. [PMID: 35788130 PMCID: PMC9253313 DOI: 10.1038/s41368-022-00185-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022] Open
Abstract
The progressive destruction of condylar cartilage is a hallmark of the temporomandibular joint (TMJ) osteoarthritis (OA); however, its mechanism is incompletely understood. Here, we show that Kindlin-2, a key focal adhesion protein, is strongly detected in cells of mandibular condylar cartilage in mice. We find that genetic ablation of Kindlin-2 in aggrecan-expressing condylar chondrocytes induces multiple spontaneous osteoarthritic lesions, including progressive cartilage loss and deformation, surface fissures, and ectopic cartilage and bone formation in TMJ. Kindlin-2 loss significantly downregulates the expression of aggrecan, Col2a1 and Proteoglycan 4 (Prg4), all anabolic extracellular matrix proteins, and promotes catabolic metabolism in TMJ cartilage by inducing expression of Runx2 and Mmp13 in condylar chondrocytes. Kindlin-2 loss decreases TMJ chondrocyte proliferation in condylar cartilages. Furthermore, Kindlin-2 loss promotes the release of cytochrome c as well as caspase 3 activation, and accelerates chondrocyte apoptosis in vitro and TMJ. Collectively, these findings reveal a crucial role of Kindlin-2 in condylar chondrocytes to maintain TMJ homeostasis.
Collapse
Affiliation(s)
- Yumei Lai
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Wei Zheng
- Department of Orthopaedic Center, Xinjiang Production and Construction Corps Hospital, Urumqi, China
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Christopher C Xiao
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Sheng Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Qinnan Yan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Peijun Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, China
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 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 Key Laboratory of Cell Microenvironment, Shenzhen, China.
| |
Collapse
|
31
|
Gomathi K, Rohini M, Vairamani M, Selvamurugan N. Identification and characterization of TGF-β1-responsive Runx2 acetylation sites for matrix Metalloproteinase-13 expression in osteoblastic cells. Biochimie 2022; 201:1-6. [PMID: 35779648 DOI: 10.1016/j.biochi.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/26/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022]
Abstract
In skeletal tissues, transforming growth factor-beta 1 (TGF-β1) serves a number of activities. For example, in osteoblastic cells, TGF-β1 stimulates the expression of matrix metalloproteinase-13 (MMP-13, a bone remodeling gene), which requires the bone transcription factor Runx2. Although TGF-β1 is known to stimulate Runx2 acetylation, the sites involved in MMP-13 gene activation remain unknown. Mass spectrometry analysis revealed that Runx2 was acetylated at one site (K134) and three sites (K24, K134, and K169) following control and TGF-β1-treatment, respectively, in osteoblastic cells. In addition, we mutated the lysine residues in the Runx2 construct into arginine and transfected the construct into mouse mesenchymal stem cells (C3H10T1/2). Wild-type Runx2 expression and acetylation were significantly increased by TGF-β1-treatment, whereas this effect was decreased in the presence of the Runx2 double mutant construct (K24 + K169) in C3H10T1/2 cells. TGF-β1 enhanced MMP-13 promoter activity in cells transfected with the wild-type Runx2 construct, but this effect was considerably reduced in cells transfected with the Runx2 double mutant construct (K24 + K169), according to a luciferase reporter test. Hence, the stability of Runx2 may be mediated by TGF-β1-induced acetylation at K24 and K169 and is required for MMP-13 expression in osteoblastic cells. These findings add to our knowledge of TGF-β1, Runx2, and MMP-13's physiological roles in bone metabolism.
Collapse
Affiliation(s)
- Kanagaraj Gomathi
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Muthukumar Rohini
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Mariappan Vairamani
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
32
|
Smith SS, Chu D, Qu T, Aggleton JA, Schneider RA. Species-specific sensitivity to TGFβ signaling and changes to the Mmp13 promoter underlie avian jaw development and evolution. eLife 2022; 11:e66005. [PMID: 35666955 PMCID: PMC9246370 DOI: 10.7554/elife.66005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/03/2022] [Indexed: 12/02/2022] Open
Abstract
Precise developmental control of jaw length is critical for survival, but underlying molecular mechanisms remain poorly understood. The jaw skeleton arises from neural crest mesenchyme (NCM), and we previously demonstrated that these progenitor cells express more bone-resorbing enzymes including Matrix metalloproteinase 13 (Mmp13) when they generate shorter jaws in quail embryos versus longer jaws in duck. Moreover, if we inhibit bone resorption or Mmp13, we can increase jaw length. In the current study, we uncover mechanisms establishing species-specific levels of Mmp13 and bone resorption. Quail show greater activation of and sensitivity to transforming growth factor beta (TGFβ) signaling than duck; where intracellular mediators like SMADs and targets like Runt-related transcription factor 2 (Runx2), which bind Mmp13, become elevated. Inhibiting TGFβ signaling decreases bone resorption, and overexpressing Mmp13 in NCM shortens the duck lower jaw. To elucidate the basis for this differential regulation, we examine the Mmp13 promoter. We discover a SMAD-binding element and single nucleotide polymorphisms (SNPs) near a RUNX2-binding element that distinguish quail from duck. Altering the SMAD site and switching the SNPs abolish TGFβ sensitivity in the quail Mmp13 promoter but make the duck promoter responsive. Thus, differential regulation of TGFβ signaling and Mmp13 promoter structure underlie avian jaw development and evolution.
Collapse
Affiliation(s)
- Spenser S Smith
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Daniel Chu
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Tiange Qu
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Jessye A Aggleton
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
33
|
Wilde C, Mitgau J, Suchý T, Schoeneberg T, Liebscher I. Translating the Force - mechano-sensing GPCRs. Am J Physiol Cell Physiol 2022; 322:C1047-C1060. [PMID: 35417266 DOI: 10.1152/ajpcell.00465.2021] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Incorporating mechanical cues into cellular responses allows us to experience our direct environment. Specialized cells can perceive and discriminate between different physical properties such as level of vibration, temperature, or pressure. Mechanical forces are abundant signals that also shape general cellular responses such as cytoskeletal rearrangement, differentiation, or migration and contribute to tissue development and function. The molecular structures that perceive and transduce mechanical forces are specialized cytoskeletal proteins, cell junction molecules, and membrane proteins such as ion channels and metabotropic receptors. G protein-coupled receptors (GPCRs) have attracted attention as metabotropic force receptors as they are among the most important drug targets. This review summarizes the function of mechano-sensitive GPCRs, specifically, the angiotensin II type 1 receptor and adrenergic, apelin, histamine, parathyroid hormone 1, and orphan receptors, focusing particularly on the advanced knowledge gained from adhesion-type GPCRs. We distinguish between shear stress and cell swelling/stretch as the two major types of mechano-activation of these receptors and contemplate the potential contribution of the force-from-lipid and force-from-tether models that have previously been suggested for ion channels.
Collapse
Affiliation(s)
- Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Jakob Mitgau
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Tomás Suchý
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Torsten Schoeneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| |
Collapse
|
34
|
López-Jiménez C, Chiu LL, Waldman SD, Guilak F, Koch TG. TRPV4 activation enhances compressive properties and glycosaminoglycan deposition of equine neocartilage sheets. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100263. [DOI: 10.1016/j.ocarto.2022.100263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 11/24/2022] Open
|
35
|
Gomathi K, Rohini M, Partridge NC, Selvamurugan N. Regulation of transforming growth factor-β1-stimulation of Runx2 acetylation for matrix metalloproteinase 13 expression in osteoblastic cells. Biol Chem 2022; 403:305-315. [PMID: 34643076 DOI: 10.1515/hsz-2021-0292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/30/2021] [Indexed: 01/12/2023]
Abstract
Transforming growth factor beta 1 (TGF-β1) functions as a coupling factor between bone development and resorption. Matrix metalloproteinase 13 (MMP13) is important in bone remodeling, and skeletal dysplasia is caused by a deficiency in MMP13 expre-ssion. Runx2, a transcription factor is essential for bone development, and MMP13 is one of its target genes. TGF-β1 promoted Runx2 phosphorylation, which was necessary for MMP13 production in osteoblastic cells, as we previously shown. Since the phosphorylation of some proteins causes them to be degraded by the ubiquitin/proteasome pathway, we hypothesized that TGF-β1 might stabilize the phosphorylated Runx2 protein for its activity by other post-translational modification (PTM). This study demonstrated that TGF-β1-stimulated Runx2 acetylation in rat osteoblastic cells. p300, a histone acetyltransferase interacted with Runx2, and it promoted Runx2 acetylation upon TGF-β1-treatment in these cells. Knockdown of p300 decreased the TGF-β1-stimulated Runx2 acetylation and MMP13 expression in rat osteoblastic cells. TGF-β1-treatment stimulated the acetylated Runx2 bound at the MMP13 promoter, and knockdown of p300 reduced this effect in these cells. Overall, our studies identified the transcriptional regulation of MMP13 by TGF-β1 via Runx2 acetylation in rat osteoblastic cells, and these findings contribute to the knowledge of events presiding bone metabolism.
Collapse
Affiliation(s)
- Kanagaraj Gomathi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Muthukumar Rohini
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Nicola C Partridge
- Department of Molecular Pathobiology, New York University College Dentistry, New York, NY, USA
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
36
|
Somemura S, Kumai T, Yatabe K, Sasaki C, Fujiya H, Niki H, Yudoh K. Physiologic Mechanical Stress Directly Induces Bone Formation by Activating Glucose Transporter 1 (Glut 1) in Osteoblasts, Inducing Signaling via NAD+-Dependent Deacetylase (Sirtuin 1) and Runt-Related Transcription Factor 2 (Runx2). Int J Mol Sci 2021; 22:9070. [PMID: 34445787 PMCID: PMC8396442 DOI: 10.3390/ijms22169070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/23/2022] Open
Abstract
Mechanical stress is an important factor affecting bone tissue homeostasis. We focused on the interactions among mechanical stress, glucose uptake via glucose transporter 1 (Glut1), and the cellular energy sensor sirtuin 1 (SIRT1) in osteoblast energy metabolism, since it has been recognized that SIRT1, an NAD+-dependent deacetylase, may function as a master regulator of the mechanical stress response as well as of cellular energy metabolism (glucose metabolism). In addition, it has already been demonstrated that SIRT1 regulates the activity of the osteogenic transcription factor runt-related transcription factor 2 (Runx2). The effects of mechanical loading on cellular activities and the expressions of Glut1, SIRT1, and Runx2 were evaluated in osteoblasts and chondrocytes in a 3D cell-collagen sponge construct. Compressive mechanical loading increased osteoblast activity. Mechanical loading also significantly increased the expression of Glut1, significantly decreased the expression of SIRT1, and significantly increased the expression of Runx2 in osteoblasts in comparison with non-loaded osteoblasts. Incubation with a Glut1 inhibitor blocked mechanical stress-induced changes in SIRT1 and Runx2 in osteoblasts. In contrast with osteoblasts, the expressions of Glut1, SIRT1, and Runx2 in chondrocytes were not affected by loading. Our present study indicated that mechanical stress induced the upregulation of Glut1 following the downregulation of SIRT1 and the upregulation of Runx2 in osteoblasts but not in chondrocytes. Since SIRT1 is known to negatively regulate Runx2 activity, a mechanical stress-induced downregulation of SIRT1 may lead to the upregulation of Runx2, resulting in osteoblast differentiation. Incubation with a Glut1 inhibitor the blocked mechanical stress-induced downregulation of SIRT1 following the upregulation of Runx2, suggesting that Glut1 is necessary to mediate the responses of SIRT1 and Runx2 to mechanical loading in osteoblasts.
Collapse
Affiliation(s)
- Shu Somemura
- Department of Sports Medicine, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8511, Japan; (S.S.); (K.Y.); (H.F.)
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan; (T.K.); (H.N.)
| | - Takanori Kumai
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan; (T.K.); (H.N.)
| | - Kanaka Yatabe
- Department of Sports Medicine, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8511, Japan; (S.S.); (K.Y.); (H.F.)
| | - Chizuko Sasaki
- Institute for Ultrastructural Morphology, St. Marianna University Graduate School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan;
| | - Hiroto Fujiya
- Department of Sports Medicine, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8511, Japan; (S.S.); (K.Y.); (H.F.)
| | - Hisateru Niki
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan; (T.K.); (H.N.)
| | - Kazuo Yudoh
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki 216-8512, Japan
| |
Collapse
|
37
|
Park S, Bello A, Arai Y, Ahn J, Kim D, Cha KY, Baek I, Park H, Lee SH. Functional Duality of Chondrocyte Hypertrophy and Biomedical Application Trends in Osteoarthritis. Pharmaceutics 2021; 13:pharmaceutics13081139. [PMID: 34452101 PMCID: PMC8400409 DOI: 10.3390/pharmaceutics13081139] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Chondrocyte hypertrophy is one of the key indicators in the progression of osteoarthritis (OA). However, compared with other OA indications, such as cartilage collapse, sclerosis, inflammation, and protease activation, the mechanisms by which chondrocyte hypertrophy contributes to OA remain elusive. As the pathological processes in the OA cartilage microenvironment, such as the alterations in the extracellular matrix, are initiated and dictated by the physiological state of the chondrocytes, in-depth knowledge of chondrocyte hypertrophy is necessary to enhance our understanding of the disease pathology and develop therapeutic agents. Chondrocyte hypertrophy is a factor that induces OA progression; it is also a crucial factor in the endochondral ossification. This review elaborates on this dual functionality of chondrocyte hypertrophy in OA progression and endochondral ossification through a description of the characteristics of various genes and signaling, their mechanism, and their distinguishable physiological effects. Chondrocyte hypertrophy in OA progression leads to a decrease in chondrogenic genes and destruction of cartilage tissue. However, in endochondral ossification, it represents an intermediate stage at the process of differentiation of chondrocytes into osteogenic cells. In addition, this review describes the current therapeutic strategies and their mechanisms, involving genes, proteins, cytokines, small molecules, three-dimensional environments, or exosomes, against the OA induced by chondrocyte hypertrophy. Finally, this review proposes that the contrasting roles of chondrocyte hypertrophy are essential for both OA progression and endochondral ossification, and that this cellular process may be targeted to develop OA therapeutics.
Collapse
Affiliation(s)
- Sunghyun Park
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea
| | - Alvin Bello
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Jinsung Ahn
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Dohyun Kim
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Kyung-Yup Cha
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Inho Baek
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
| | - Hansoo Park
- School of Integrative Engineering, Chung-ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea;
| | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University-Seoul, Seoul 04620, Korea; (S.P.); (A.B.); (Y.A.); (J.A.); (D.K.); (K.-Y.C.); (I.B.)
- Correspondence: ; Tel.: +82-31-961-5153; Fax: +82-31-961-5108
| |
Collapse
|
38
|
Wen ZH, Huang JS, Lin YY, Yao ZK, Lai YC, Chen WF, Liu HT, Lin SC, Tsai YC, Tsai TC, Jean YH. Chondroprotective Effects of a Histone Deacetylase Inhibitor, Panobinostat, on Pain Behavior and Cartilage Degradation in Anterior Cruciate Ligament Transection-Induced Experimental Osteoarthritic Rats. Int J Mol Sci 2021; 22:ijms22147290. [PMID: 34298911 PMCID: PMC8306086 DOI: 10.3390/ijms22147290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is the most common articular degenerative disease characterized by chronic pain, joint inflammation, and movement limitations, which are significantly influenced by aberrant epigenetic modifications of numerous OA-susceptible genes. Recent studies revealed that both the abnormal activation and differential expression of histone deacetylases (HDACs) might contribute to OA pathogenesis. In this study, we investigated the chondroprotective effects of a marine-derived HDAC inhibitor, panobinostat, on anterior cruciate ligament transection (ACLT)-induced experimental OA rats. The intra-articular administration of 2 or 10 µg of panobinostat (each group, n = 7) per week from the 6th to 17th week attenuates ACLT-induced nociceptive behaviors, including secondary mechanical allodynia and weight-bearing distribution. Histopathological and microcomputed tomography analysis showed that panobinostat significantly prevents cartilage degeneration after ACLT. Moreover, intra-articular panobinostat exerts hypertrophic effects in the chondrocytes of articular cartilage by regulating the protein expressions of HDAC4, HDAC6, HDAC7, runt-domain transcription factor-2, and matrix metalloproteinase-13. The study indicated that HDACs might have different modulations on the chondrocyte phenotype in the early stages of OA development. These results provide new evidence that panobinostat may be a potential therapeutic drug for OA.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (Z.-H.W.); (Z.-K.Y.); (Y.-C.L.); (W.-F.C.)
| | - Jhy-Shrian Huang
- Section of Orthopedics, Department of Surgery, Antai Medical Care Corporation Anti Tian-Sheng Memorial Hospital, PingTong 92842, Taiwan;
| | - Yen-You Lin
- Department of Sports Medicine, China Medical University, No. 91 Hsueh-Shih Road, Taichung 40402, Taiwan;
| | - Zhi-Kang Yao
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (Z.-H.W.); (Z.-K.Y.); (Y.-C.L.); (W.-F.C.)
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung 81341, Taiwan
| | - Yu-Cheng Lai
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (Z.-H.W.); (Z.-K.Y.); (Y.-C.L.); (W.-F.C.)
- Department of Orthopedics, Asia University Hospital, Taichung 41354, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (Z.-H.W.); (Z.-K.Y.); (Y.-C.L.); (W.-F.C.)
- Department of Neurosurgery, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 83301, Taiwan
| | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan;
| | - Sung-Chun Lin
- Department of Orthopedic Surgery, Pingtung Christian Hospital, No. 60 Dalian Road, Pingtung 90059, Taiwan;
| | - Yu-Chi Tsai
- National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan;
| | - Tsung-Chang Tsai
- Section of Nephrology, Department of Medicine, Antai Medical Care Corporation Anti Tian-Sheng Memorial Hospital, Pingtung 92842, Taiwan;
| | - Yen-Hsuan Jean
- Section of Orthopedics, Department of Surgery, Antai Medical Care Corporation Anti Tian-Sheng Memorial Hospital, PingTong 92842, Taiwan;
- Correspondence: ; Tel.: +886-8-8329966
| |
Collapse
|
39
|
Li X, Dai B, Guo J, Zheng L, Guo Q, Peng J, Xu J, Qin L. Nanoparticle-Cartilage Interaction: Pathology-Based Intra-articular Drug Delivery for Osteoarthritis Therapy. NANO-MICRO LETTERS 2021; 13:149. [PMID: 34160733 PMCID: PMC8222488 DOI: 10.1007/s40820-021-00670-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 05/03/2023]
Abstract
Osteoarthritis is the most prevalent chronic and debilitating joint disease, resulting in huge medical and socioeconomic burdens. Intra-articular administration of agents is clinically used for pain management. However, the effectiveness is inapparent caused by the rapid clearance of agents. To overcome this issue, nanoparticles as delivery systems hold considerable promise for local control of the pharmacokinetics of therapeutic agents. Given the therapeutic programs are inseparable from pathological progress of osteoarthritis, an ideal delivery system should allow the release of therapeutic agents upon specific features of disorders. In this review, we firstly introduce the pathological features of osteoarthritis and the design concept for accurate localization within cartilage for sustained drug release. Then, we review the interactions of nanoparticles with cartilage microenvironment and the rational design. Furthermore, we highlight advances in the therapeutic schemes according to the pathology signals. Finally, armed with an updated understanding of the pathological mechanisms, we place an emphasis on the development of "smart" bioresponsive and multiple modality nanoparticles on the near horizon to interact with the pathological signals. We anticipate that the exploration of nanoparticles by balancing the efficacy, safety, and complexity will lay down a solid foundation tangible for clinical translation.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China
| | - Quanyi Guo
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
40
|
He F, Matsumoto Y, Asano Y, Yamamura Y, Katsuyama T, La Rose J, Tomonobu N, Komalasari NLGY, Sakaguchi M, Rottapel R, Wada J. RUNX2 Phosphorylation by Tyrosine Kinase ABL Promotes Breast Cancer Invasion. Front Oncol 2021; 11:665273. [PMID: 34136397 PMCID: PMC8201617 DOI: 10.3389/fonc.2021.665273] [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: 02/07/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022] Open
Abstract
Activity of transcription factors is normally regulated through interaction with other transcription factors, chromatin remodeling proteins and transcriptional co-activators. In distinction to these well-established transcriptional controls of gene expression, we have uncovered a unique activation model of transcription factors between tyrosine kinase ABL and RUNX2, an osteoblastic master transcription factor, for cancer invasion. We show that ABL directly binds to, phosphorylates, and activates RUNX2 through its SH2 domain in a kinase activity-dependent manner and that the complex formation of these proteins is required for expression of its target gene MMP13. Additionally, we show that the RUNX2 transcriptional activity is dependent on the number of its tyrosine residues that are phosphorylated by ABL. In addition to regulation of RUNX2 activity, we show that ABL transcriptionally enhances RUNX2 expression through activation of the bone morphogenetic protein (BMP)-SMAD pathway. Lastly, we show that ABL expression in highly metastatic breast cancer MDA-MB231 cells is associated with their invasive capacity and that ABL-mediated invasion is abolished by depletion of endogenous RUNX2 or MMP13. Our genetic and biochemical evidence obtained in this study contributes to a mechanistic insight linking ABL-mediated phosphorylation and activation of RUNX2 to induction of MMP13, which underlies a fundamental invasive capacity in cancer and is different from the previously described model of transcriptional activation.
Collapse
Affiliation(s)
- Fang He
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yosuke Asano
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuriko Yamamura
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Katsuyama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jose La Rose
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ni Luh Gede Yoni Komalasari
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Robert Rottapel
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
41
|
Chen H, Tan XN, Hu S, Liu RQ, Peng LH, Li YM, Wu P. Molecular Mechanisms of Chondrocyte Proliferation and Differentiation. Front Cell Dev Biol 2021; 9:664168. [PMID: 34124045 PMCID: PMC8194090 DOI: 10.3389/fcell.2021.664168] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/20/2022] Open
Abstract
Cartilage is a kind of connective tissue that buffers pressure and is essential to protect joint movement. It is difficult to self-recover once cartilage is damaged due to the lack of blood vessels, lymph, and nerve tissues. Repair of cartilage injury is mainly achieved by stimulating chondrocyte proliferation and extracellular matrix (ECM) synthesis. Cartilage homeostasis involves the regulation of multiple growth factors and the transduction of cellular signals. It is a very complicated process that has not been elucidated in detail. In this review, we summarized a variety of signaling molecules related to chondrocytes function. Especially, we described the correlation between chondrocyte-specific regulatory factors and cell signaling molecules. It has potential significance for guiding the treatment of cartilage injury.
Collapse
Affiliation(s)
- Hui Chen
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| | - Xiao-Ning Tan
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China
| | - Shi Hu
- Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China.,Center for Bionic Sensing and Intelligence, Institute of Bio-medical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ren-Qin Liu
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| | - Li-Hong Peng
- School of Computer, Hunan University of Technology, Zhuzhou, China
| | - Yong-Min Li
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China
| | - Ping Wu
- Hunan University of Chinese Medicine & Hunan Academy of Chinese Medicine, Changsha, China.,The Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China.,Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| |
Collapse
|
42
|
Li B, Guan G, Mei L, Jiao K, Li H. Pathological mechanism of chondrocytes and the surrounding environment during osteoarthritis of temporomandibular joint. J Cell Mol Med 2021; 25:4902-4911. [PMID: 33949768 PMCID: PMC8178251 DOI: 10.1111/jcmm.16514] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Temporomandibular joint (TMJ) osteoarthritis is a common chronic degenerative disease of the TMJ. In order to explore its aetiology and pathological mechanism, many animal models and cell models have been constructed to simulate the pathological process of TMJ osteoarthritis. The main pathological features of TMJ osteoarthritis include chondrocyte death, extracellular matrix (ECM) degradation and subchondral bone remodelling. Chondrocyte apoptosis accelerates the destruction of cartilage. However, autophagy has a protective effect on condylar chondrocytes. Degradation of ECM not only changes the properties of cartilage but also affects the phenotype of chondrocytes. The loss of subchondral bone in the early stages of TMJ osteoarthritis plays an aetiological role in the onset of osteoarthritis. In recent years, increasing evidence has suggested that chondrocyte hypertrophy and endochondral angiogenesis promote TMJ osteoarthritis. Hypertrophic chondrocytes secrete many factors that promote cartilage degeneration. These chondrocytes can further differentiate into osteoblasts and osteocytes and accelerate cartilage ossification. Intrachondral angiogenesis and neoneurogenesis are considered to be important triggers of arthralgia in TMJ osteoarthritis. Many molecular signalling pathways in endochondral osteogenesis are responsible for TMJ osteoarthritis. These latest discoveries in TMJ osteoarthritis have further enhanced the understanding of this disease and contributed to the development of molecular therapies. This paper summarizes recent cognition on the pathogenesis of TMJ osteoarthritis, focusing on the role of chondrocyte hypertrophy degeneration and cartilage angiogenesis.
Collapse
Affiliation(s)
- Baochao Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guangzhao Guan
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Li Mei
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Kai Jiao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
43
|
Cryptic ligand on collagen matrix unveiled by MMP13 accelerates bone tissue regeneration via MMP13/Integrin α3/RUNX2 feedback loop. Acta Biomater 2021; 125:219-230. [PMID: 33677160 DOI: 10.1016/j.actbio.2021.02.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) remodeling is necessary for the development and self-healing of tissue, and the process is tissue specific. Matrix metalloproteinases (MMPs) play a role in ECM remodeling by unwinding and cleaving ECM. We hypothesized that ECM remodeling by MMPs is involved in the differentiation of stem cells into specific lineages during self-healing. To prove the hypothesis, we investigated which MMPs are involved in the osteogenic differentiation of human mesenchymal stem cells (hMSCs) grown on a type I collagen (Col I) matrix, and we found that specifically high expression of MMP13 in hMSCs grown on a Col I matirx during osteogenic differentiation. Moreover, knocking down of MMP13 decreased the osteogenic differentiation of hMSCs grown on a Col I matrix. In addition, pre-treatment of recombinant human MMP13 lead to remodeling of Col I matrix and increased the osteogenic differentiation of hMSCs and in vivo bone formation following the upregulation of the expression of runt-related transcription factor 2 (RUNX2), integrin α3 (ITGA3), and focal adhesion kinase. Furthermore, the transcription factor RUNX2 bound to the MMP13 promoter. These results suggest that growth on a remodeled Col I matrix by MMP13 stimulates osteogenic differentiation of hMSCs and self-healing of bone tissue via an MMP13/ITGA3/RUNX2 positive feedback loop. STATEMENT OF SIGNIFICANCE: Self-healing of tissue could be the key to treating diseases that cannot be overcome by present technology. We investigated the mechanism underlying the self-healing of tissue and we found that the osteogenic differentiation was increased in hMSCs grown on a remodeled Col I matrix by the optimized concentration of MMP13 not in hMSCs grown on a Col I fragments cleaved by a high concentration of MMP13. In addition, we found the remodeled Col I matrix by MMP13 increased the osteogenic capacity through a MMP13/integrin α3/RUNX2 positive feedback loop. This result would be able to not only provide a strategy for bone tissue-specific functional materials following strong evidence about the self-healing mechanism of bone through the interaction between stem cells and the ECM matrix. As such, we strongly believe our finding will be of interest to researchers studying biomaterials, stem cell biology and matrix interaction for regenerative medicine and therapy.
Collapse
|
44
|
McClurg O, Tinson R, Troeberg L. Targeting Cartilage Degradation in Osteoarthritis. Pharmaceuticals (Basel) 2021; 14:ph14020126. [PMID: 33562742 PMCID: PMC7916085 DOI: 10.3390/ph14020126] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis is a common, degenerative joint disease with significant socio-economic impact worldwide. There are currently no disease-modifying drugs available to treat the disease, making this an important area of pharmaceutical research. In this review, we assessed approaches being explored to directly inhibit metalloproteinase-mediated cartilage degradation and to counteract cartilage damage by promoting growth factor-driven repair. Metalloproteinase-blocking antibodies are discussed, along with recent clinical trials on FGF18 and Wnt pathway inhibitors. We also considered dendrimer-based approaches being developed to deliver and retain such therapeutics in the joint environment. These may reduce systemic side effects while improving local half-life and concentration. Development of such targeted anabolic therapies would be of great benefit in the osteoarthritis field.
Collapse
|
45
|
Song H, Du H, Li J, Wang M, Wang J, Ju X, Mu W. Effect of fibroblast growth factor 2 on degenerative endplate chondrocyte: From anabolism to catabolism. Exp Mol Pathol 2020; 118:104590. [PMID: 33285208 DOI: 10.1016/j.yexmp.2020.104590] [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: 06/11/2020] [Revised: 10/30/2020] [Accepted: 12/01/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Endplate degeneration is characterized by an unbalance between the anabolism and catabolism of endplate chondrocyte (CH). Fibroblast growth factor 2 (FGF2) has been shown to promote cartilage repair by increasing articular CH anabolic activity. We aimed to explore the effect of FGF2 on the metabolism of endplate CH to elucidate whether FGF2 could be used as a therapy to delay the endplate degeneration. METHODS We collected the endplate tissue from the patients and tested the collagen II mRNA level as the anabolic marker and the MMP-13 and TIMP-4 expression as the catabolic markers. The FGF2, FGF receptor 1 (FGFR1), and FGFR3 mRNA expression of the endplate tissue were also analyzed. Besides, we treated the CHs with exogenic FGF2 protein, measured the markers mentioned above, the proliferation and the apoptosis of the CHs. To compare the effect of FGF2 on the CHs with or without degeneration, we also induced CHs degeneration by interleukin-1β (IL-1β) stimulation and used the FGF2 protein to treat the degenerative CHs. RESULTS Severely degenerative endplate had a lower collagen II and TIMP-4 mRNA level, but it expressed a more massive amount of MMP-13, FGF2, and FGFR1. FGF2 supplement upregulated the FGFR1/FGFR3, TIMP-4, collagen II expression, and promoted the CHs proliferation. In the first 24 h of IL-1β treatment, the FGF2 mRNA expression was suppressed, but it significantly increased 48 h later. Meanwhile, the FGFR1 was upregulated, and FGFR3 was inhibited by IL-1β treatment. Interestingly, the FGF2 protein supplement accelerated the degenerative CHs catabolism by decreasing collagen II and TIPM-4 expression but increasing MMP-13. However, the FGF2 could promote the anabolism process in case of the blocking of FGFR1. The FGF2 supplement could also promote the proliferation and inhibited the apoptosis of degenerative CHs, which could be magnified by FGFR1 blocking. CONCLUSIONS The results demonstrate that FGF2 is upregulated in the highly degenerative endplate. The supplement of FGF2 contributes to the anabolism in the early phase of endplate degeneration. In the later stage of endplate degeneration, FGF2 turns to accelerate the catabolism, which can be rejected by the reasonable use of FGFR1 inhibitors.
Collapse
Affiliation(s)
- Hua Song
- School of Medicine, Shandong University, Jinan 250012, China
| | - Hongyang Du
- Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou 277500, China
| | - Jie Li
- Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou 277500, China
| | - Mingming Wang
- Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou 277500, China
| | - Jianhua Wang
- Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou 277500, China
| | - Xuezhuang Ju
- Department of Orthopaedics, Tengzhou Central People's Hospital, Tengzhou 277500, China
| | - Weidong Mu
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.
| |
Collapse
|
46
|
Li JX, He JJ, Elsheikha HM, Ma J, Xu XP, Zhu XQ. ROP18-Mediated Transcriptional Reprogramming of HEK293T Cell Reveals New Roles of ROP18 in the Interplay Between Toxoplasma gondii and the Host Cell. Front Cell Infect Microbiol 2020; 10:586946. [PMID: 33330132 PMCID: PMC7734210 DOI: 10.3389/fcimb.2020.586946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/30/2020] [Indexed: 12/02/2022] Open
Abstract
Toxoplasma gondii secretes a number of virulence-related effector proteins, such as the rhoptry protein 18 (ROP18). To further broaden our understanding of the molecular functions of ROP18, we examined the transcriptional response of human embryonic kidney cells (HEK293T) to ROP18 of type I T. gondii RH strain. Using RNA-sequencing, we compared the transcriptome of ROP18-expressing HEK293T cells to control HEK293T cells. Our analysis revealed that ROP18 altered the expression of 750 genes (467 upregulated genes and 283 downregulated genes) in HEK293T cells. Gene ontology (GO) and pathway enrichment analyses showed that differentially expressed genes (DEGs) were significantly enriched in extracellular matrix– and immune–related GO terms and pathways. KEGG pathway enrichment analysis revealed that DEGs were involved in several disease-related pathways, such as nervous system diseases and eye disease. ROP18 significantly increased the alternative splicing pattern “retained intron” and altered the expression of 144 transcription factors (TFs). These results provide new insight into how ROP18 may influence biological processes in the host cells via altering the expression of genes, TFs, and pathways. More in vitro and in vivo studies are required to substantiate these findings.
Collapse
Affiliation(s)
- Jie-Xi Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Jun Ma
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiao-Pei Xu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Heilongjiang Key Laboratory for Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| |
Collapse
|
47
|
Bone phenotypes in rheumatology - there is more to bone than just bone. BMC Musculoskelet Disord 2020; 21:789. [PMID: 33248451 PMCID: PMC7700716 DOI: 10.1186/s12891-020-03804-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis, rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis, all have one clear common denominator; an altered turnover of bone. However, this may be more complex than a simple change in bone matrix and mineral turnover. While these diseases share a common tissue axis, their manifestations in the area of pathology are highly diverse, ranging from sclerosis to erosion of bone in different regions. The management of these diseases will benefit from a deeper understanding of the local versus systemic effects, the relation to the equilibrium of the bone balance (i.e., bone formation versus bone resorption), and the physiological and pathophysiological phenotypes of the cells involved (e.g., osteoblasts, osteoclasts, osteocytes and chondrocytes). For example, the process of endochondral bone formation in chondrocytes occurs exists during skeletal development and healthy conditions, but also in pathological conditions. This review focuses on the complex molecular and cellular taxonomy of bone in the context of rheumatological diseases that alter bone matrix composition and maintenance, giving rise to different bone turnover phenotypes, and how biomarkers (biochemical markers) can be applied to potentially describe specific bone phenotypic tissue profiles.
Collapse
|
48
|
Jahangir S, Eglin D, Pötter N, Khozaei Ravari M, Stoddart MJ, Samadikuchaksaraei A, Alini M, Baghaban Eslaminejad M, Safa M. Inhibition of hypertrophy and improving chondrocyte differentiation by MMP-13 inhibitor small molecule encapsulated in alginate-chondroitin sulfate-platelet lysate hydrogel. Stem Cell Res Ther 2020; 11:436. [PMID: 33036643 PMCID: PMC7545577 DOI: 10.1186/s13287-020-01930-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/08/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells are a promising cell source for chondrogenic differentiation and have been widely used in several preclinical and clinical studies. However, they are prone to an unwanted differentiation process towards hypertrophy that limits their therapeutic efficacy. Matrix metallopeptidase 13 (MMP-13) is a well-known factor regulated during this undesirable event. MMP-13 is a collagen degrading enzyme, which is also highly expressed in the hypertrophic zone of the growth plate and in OA cartilage. Accordingly, we investigated the effect of MMP-13 inhibition on MSC hypertrophy. METHODS In this study, 5-bromoindole-2-carboxylic acid (BICA) was used as an inhibitory agent for MMP-13 expression. After identifying its optimal concentration, BICA was mixed into a hydrogel and the release rate was studied. To prepare the ideal hydrogel, chondroitin sulfate (CS) and platelet lysate (PL) were mixed with sodium alginate (Alg) at concentrations selected based on synergistic mechanical and rheometric properties. Then, four hydrogels were prepared by combining alginate (1.5%w/v) and/or CS (1%w/v) and/or PL (20%v/v). The chondrogenic potential and progression to hypertrophy of human bone marrow-derived mesenchymal stem cell (hBM-MSC)-loaded hydrogels were investigated under free swelling and mechanical loading conditions, in the presence and absence of BICA. RESULTS Viability of hBM-MSCs seeded in the four hydrogels was similar. qRT-PCR revealed that BICA could successfully inhibit MMP-13 expression, which led to an inhibition of Coll X and induction of Coll-II, in both free swelling and loading conditions. The GAG deposition was higher in the group combining BICA and mechanical stimulation. CONCLUSIONS It is concluded that BICA inhibition of MMP-13 reduces MSC hypertrophy during chondrogenesis.
Collapse
Affiliation(s)
- Shahrbanoo Jahangir
- Department of Tissue engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - David Eglin
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Naomi Pötter
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
- Department of orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center Albert-Ludwigs University, Albert-Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Mojtaba Khozaei Ravari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
- Department of orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center Albert-Ludwigs University, Albert-Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Ali Samadikuchaksaraei
- Department of Tissue engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mauro Alini
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland.
| | - Mohammadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Majid Safa
- Department of Tissue engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Vinod E, Kachroo U, Rebekah G, Yadav BK, Ramasamy B. Characterization of human articular chondrocytes and chondroprogenitors derived from non-diseased and osteoarthritic knee joints to assess superiority for cell-based therapy. Acta Histochem 2020; 122:151588. [PMID: 32778244 DOI: 10.1016/j.acthis.2020.151588] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/27/2020] [Accepted: 06/27/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Cell based therapy is constantly underway since regeneration of genuine hyaline cartilage is under par. Much attention has been afforded to chondroprogenitors recently, as an alternative cell substitute for cartilage repair. Although single source derivation of chondrocytes and chondroprogenitors is advantageous, lack of a characteristic differentiating marker obscures clear identification, which is essential to create a biological profile and is also required to assess cell type superiority for cartilage repair. METHODS Cells obtained from three non-diseased/osteoarthritic human knee joints each, were expanded in culture up to passage 10. Characterization studies were performed using flow cytometry; gene expression was studied using RT-PCR; growth kinetics and tri-lineage differentiation was also studied to construct a better profile of chondroprogenitors as well as chondrocytes. RESULTS AND CONCLUSION Our results showed that both cell populations exhibited similar cell surface characteristics except for non-diseased chondroprogenitors, which showed markedly low expression of CD34 and high expression of CD166. Trilineage data was suggestive of multilineage potential for both cell types with chondroprogenitors showing notably higher glycosaminoglycan and lower calcified matrix deposition. Data acquired from this study aided in describing cellular behavior of human articular cartilage derived chondroprogenitors in conditions not reported earlier. Our comparative analysis suggests that sorting based on a combination of markers (CD34- and CD166+) would yield a population of cells with minimal contamination by chondrocytes, which may provide translatable results in terms of enhanced chondrogenesis and reduced hypertrophy; both indispensable for the field of cartilage regeneration.
Collapse
Affiliation(s)
- Elizabeth Vinod
- Department of Physiology, Christian Medical College, Vellore, 632002, India; Centre for Stem Cell Research, Christian Medical College, Vellore, 632002, India.
| | - Upasana Kachroo
- Department of Physiology, Christian Medical College, Vellore, 632002, India.
| | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, 632002, India.
| | - Bijesh Kumar Yadav
- Department of Biostatistics, Christian Medical College, Vellore, 632002, India.
| | - Boopalan Ramasamy
- Centre for Stem Cell Research, Christian Medical College, Vellore, 632002, India; Department of Orthopaedics, Christian Medical College, Vellore, 632004, India.
| |
Collapse
|
50
|
Hojo H, Ohba S. Gene regulatory landscape in osteoblast differentiation. Bone 2020; 137:115458. [PMID: 32474244 DOI: 10.1016/j.bone.2020.115458] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/25/2020] [Indexed: 12/29/2022]
Abstract
The development of osteoblasts, a bone-forming cell population, occurs in conjunction with development of the skeleton, which creates our physical framework and shapes the body. In the past two decades, genetic studies have uncovered the molecular framework of this process-namely, transcriptional regulators and signaling pathways coordinate the cell fate determination and differentiation of osteoblasts in a spatial and temporal manner. Recently emerging genome-wide studies provide additional layers of understanding of the gene regulatory landscape during osteoblast differentiation, allowing us to gain novel insight into the modes of action of the key regulators, functional interaction among the regulator-bound enhancers, epigenetic regulations, and the complex nature of regulatory inputs. In this review, we summarize current understanding of the transcriptional regulation in osteoblasts, in terms of the gene regulatory landscape.
Collapse
Affiliation(s)
- Hironori Hojo
- Department of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan.
| |
Collapse
|