1
|
Hyland V, Iovine MK. Ccn2a acts downstream of cx43 to influence joint formation during zebrafish fin regeneration. Biol Open 2025; 14:bio061674. [PMID: 39963716 PMCID: PMC11876838 DOI: 10.1242/bio.061674] [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: 07/31/2024] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
This study provides new insights into the molecular pathways dictating skeletal patterning during zebrafish fin regeneration. Connexin43 (Cx43) is known to influence skeletal patterning by inhibiting evx1 expression and thereby regulating the timing of joint formation. Here, we demonstrate that cellular communication network factor 2 (ccn2a) also contributes to this pathway. We find that Ccn2a appears to act downstream of Cx43 and similarly inhibits joint formation by inhibiting evx1 expression. Pharmacological inhibition of β-catenin demonstrates that ccn2a is likely regulated by β-catenin. Additionally, this paper provides evidence that Yap signaling contributes to joint formation through regulating ccn2a. These findings provide novel insights into the role of Ccn2a during skeletal patterning.
Collapse
Affiliation(s)
- Victoria Hyland
- Lehigh University, Department of Biological Sciences, Bethlehem, PA 18015,USA
| | - M. Kathryn Iovine
- Lehigh University, Department of Biological Sciences, Bethlehem, PA 18015,USA
| |
Collapse
|
2
|
Hridayanka KSN, Duttaroy AK, Basak S. Bioactive Compounds and Their Chondroprotective Effects for Osteoarthritis Amelioration: A Focus on Nanotherapeutic Strategies, Epigenetic Modifications, and Gut Microbiota. Nutrients 2024; 16:3587. [PMID: 39519419 PMCID: PMC11547880 DOI: 10.3390/nu16213587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
In degenerative joint disease like osteoarthritis (OA), bioactive compounds like resveratrol, epigallocatechin gallate, curcumin, and other polyphenols often target various signalling pathways, including NFκB, TGFβ, and Wnt/β-catenin by executing epigenetic-modifying activities. Epigenetic modulation can target genes of disease pathophysiology via histone modification, promoter DNA methylation, and non-coding RNA expression, some of which are directly involved in OA but have been less explored. OA patients often seek options that can improve the quality of their life in addition to existing treatment with nonsteroidal anti-inflammatory drugs (NSAIDs). Although bioactive and natural compounds exhibit therapeutic potential against OA, several disadvantages loom, like insolubility and poor bioavailability. Nanoformulated bioactive compounds promise a better way to alleviate OA since they also control systemic events, including metabolic, immunological, and inflammatory responses, by modulating host gut microbiota that can regulate OA pathogenesis. Recent data suggest gut dysbiosis in OA. However, limited evidence is available on the role of bioactive compounds as epigenetic and gut modulators in ameliorating OA. Moreover, it is not known whether the effects of polyphenolic bioactive compounds on gut microbial response are mediated by epigenetic modulatory activities in OA. This narrative review highlights the nanotherapeutic strategies utilizing bioactive compounds, reporting their effects on chondrocyte growth, metabolism, and epigenetic modifications in osteoarthritis amelioration.
Collapse
Affiliation(s)
- Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway;
| | - Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| |
Collapse
|
3
|
Yang X, Liu H, Cheng S, Pan C, Cai Q, Chu X, Shi S, Wei W, He D, Cheng B, Wen Y, Jia Y, Tinkov AA, Skalny AV, Zhang F. Potential involvement of connective tissue growth factor in chondrocytes apoptosis of Kashin-Beck disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117148. [PMID: 39369662 DOI: 10.1016/j.ecoenv.2024.117148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Kashin-Beck disease (KBD) is an endemic osteoarthropathy characterized by excessive chondrocytes apoptosis. T-2 toxin exposure has been proved to be its etiology. Connective tissue growth factor (CTGF) exerts a profound influence on cartilage growth and metabolism. We investigated the potential role of CTGF in KBD development and examined CTGF alterations under T-2 toxin stimulation. METHODS The levels of CTGF and chondrocyte apoptosis-related markers in cartilage and primary chondrocytes from KBD and control groups were measured using qRT-PCR, Western blotting, immunohistochemistry, and immunofluorescence. We analyzed expression changes of these genes in response to T-2 toxin. Apoptosis rates of chondrocytes induced by T-2 toxin were measured by flow cytometry and TUNEL assay. The active pharmaceutical ingredient targeting CTGF was screened through Comparative Toxicogenomics Database, and molecular docking was performed using AutoDock Tools. RESULTS The CTGF levels in KBD cartilage and chondrocytes were significantly elevated and positively associated with the levels of apoptosis-related genes. T-2 toxin exposure increased CTGF and apoptosis-related gene levels in chondrocytes, with apoptosis rates rising alongside T-2 toxin concentration. Curcumin was identified as targeting CTGF and exhibited effective binding. It could down-regulate CTGF, apoptosis-related genes, such as Cleaved caspase 3 and BAX, and also significantly reduce apoptosis rate in chondrocytes treated with T-2 toxin. CONCLUSION CTGF plays a crucial role in the development of KBD. Curcumin has shown potential in inhibiting CTGF levels and reducing chondrocyte apoptosis, highlighting its promise as a therapeutic agent for preventing cartilage damage in KBD. Our findings provided valuable insights into the pathogenesis of KBD and could promote the development of novel therapeutic strategies for this debilitating disease.
Collapse
Affiliation(s)
- Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Huan Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Qingqing Cai
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoge Chu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Sirong Shi
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Wenming Wei
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Dan He
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Alexey A Tinkov
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow 119146, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl 150000, Russia
| | - Anatoly V Skalny
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow 119146, Russia; Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Damerau A, Rosenow E, Alkhoury D, Buttgereit F, Gaber T. Fibrotic pathways and fibroblast-like synoviocyte phenotypes in osteoarthritis. Front Immunol 2024; 15:1385006. [PMID: 38895122 PMCID: PMC11183113 DOI: 10.3389/fimmu.2024.1385006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.
Collapse
Affiliation(s)
- Alexandra Damerau
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Emely Rosenow
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| |
Collapse
|
5
|
Zhao Z, Sun X, Tu P, Ma Y, Guo Y, Zhang Y, Liu M, Wang L, Chen X, Si L, Li G, Pan Y. Mechanisms of vascular invasion after cartilage injury and potential engineering cartilage treatment strategies. FASEB J 2024; 38:e23559. [PMID: 38502020 DOI: 10.1096/fj.202302391rr] [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: 11/21/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/20/2024]
Abstract
Articular cartilage injury is one of the most common diseases in orthopedic clinics. Following an articular cartilage injury, an inability to resist vascular invasion can result in cartilage calcification by newly formed blood vessels. This process ultimately leads to the loss of joint function, significantly impacting the patient's quality of life. As a result, developing anti-angiogenic methods to repair damaged cartilage has become a popular research topic. Despite this, tissue engineering, as an anti-angiogenic strategy in cartilage injury repair, has not yet been adequately investigated. This exhaustive literature review mainly focused on the process and mechanism of vascular invasion in articular cartilage injury repair and summarized the major regulatory factors and signaling pathways affecting angiogenesis in the process of cartilage injury. We aimed to discuss several potential methods for engineering cartilage repair with anti-angiogenic strategies. Three anti-angiogenic tissue engineering methods were identified, including administering angiogenesis inhibitors, applying scaffolds to manage angiogenesis, and utilizing in vitro bioreactors to enhance the therapeutic properties of cultured chondrocytes. The advantages and disadvantages of each strategy were also analyzed. By exploring these anti-angiogenic tissue engineering methods, we hope to provide guidance for researchers in related fields for future research and development in cartilage repair.
Collapse
Affiliation(s)
- Zitong Zhao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoxian Sun
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Pengcheng Tu
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Yong Ma
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Yang Guo
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Mengmin Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Lining Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xinyu Chen
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Lin Si
- Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Guangguang Li
- Orthopedics and traumatology department, Yixing Traditional Chinese Medicine Hospital, Yixing, P.R. China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| |
Collapse
|
6
|
Brylka LJ, Alimy AR, Tschaffon-Müller MEA, Jiang S, Ballhause TM, Baranowsky A, von Kroge S, Delsmann J, Pawlus E, Eghbalian K, Püschel K, Schoppa A, Haffner-Luntzer M, Beech DJ, Beil FT, Amling M, Keller J, Ignatius A, Yorgan TA, Rolvien T, Schinke T. Piezo1 expression in chondrocytes controls endochondral ossification and osteoarthritis development. Bone Res 2024; 12:12. [PMID: 38395992 PMCID: PMC10891122 DOI: 10.1038/s41413-024-00315-x] [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: 07/18/2023] [Revised: 12/18/2023] [Accepted: 01/01/2024] [Indexed: 02/25/2024] Open
Abstract
Piezo proteins are mechanically activated ion channels, which are required for mechanosensing functions in a variety of cell types. While we and others have previously demonstrated that the expression of Piezo1 in osteoblast lineage cells is essential for bone-anabolic processes, there was only suggestive evidence indicating a role of Piezo1 and/or Piezo2 in cartilage. Here we addressed the question if and how chondrocyte expression of the mechanosensitive proteins Piezo1 or Piezo2 controls physiological endochondral ossification and pathological osteoarthritis (OA) development. Mice with chondrocyte-specific inactivation of Piezo1 (Piezo1Col2a1Cre), but not of Piezo2, developed a near absence of trabecular bone below the chondrogenic growth plate postnatally. Moreover, all Piezo1Col2a1Cre animals displayed multiple fractures of rib bones at 7 days of age, which were located close to the growth plates. While skeletal growth was only mildly affected in these mice, OA pathologies were markedly less pronounced compared to littermate controls at 60 weeks of age. Likewise, when OA was induced by anterior cruciate ligament transection, only the chondrocyte inactivation of Piezo1, not of Piezo2, resulted in attenuated articular cartilage degeneration. Importantly, osteophyte formation and maturation were also reduced in Piezo1Col2a1Cre mice. We further observed increased Piezo1 protein abundance in cartilaginous zones of human osteophytes. Finally, we identified Ptgs2 and Ccn2 as potentially relevant Piezo1 downstream genes in chondrocytes. Collectively, our data do not only demonstrate that Piezo1 is a critical regulator of physiological and pathological endochondral ossification processes, but also suggest that Piezo1 antagonists may be established as a novel approach to limit osteophyte formation in OA.
Collapse
Affiliation(s)
- Laura J Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Assil-Ramin Alimy
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Miriam E A Tschaffon-Müller
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Baden-Württemberg, 89081, Ulm, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tobias Malte Ballhause
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Julian Delsmann
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Eva Pawlus
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Kian Eghbalian
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Klaus Püschel
- Department Legal Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Baden-Württemberg, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Baden-Württemberg, 89081, Ulm, Germany
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, LS2 9JT, Leeds, UK
| | - Frank Timo Beil
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Baden-Württemberg, 89081, Ulm, Germany
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tim Rolvien
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
7
|
Xiao P, Zhu Y, Xu H, Li J, Tao A, Wang H, Cheng D, Dou X, Guo L. CTGF regulates mineralization in human mature chondrocyte by controlling Pit-1 and modulating ANK via the BMP/Smad signalling. Cytokine 2024; 174:156460. [PMID: 38134555 DOI: 10.1016/j.cyto.2023.156460] [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: 07/26/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVE Connective tissue growth factor (CTGF) exhibits potent proliferative, differentiated, and mineralizing effects, and is believed to be contribute to cartilage mineralization in Osteoarthritis (OA). However, the underlying mechanism of chondrocyte mineralization induced by CTGF remains obscure. As a key regulator of mineral responses, type III phosphate transporter 1 (Pit-1) has been associated with the pathogenesis of articular mineralization. Therefore, the primary objective of this study was to investigate whether CTGF influences the development of mature chondrocyte mineralization and the underlying mechanisms governing such mineralization. METHODS The effect of Connective tissue growth factor (CTGF) on human C-28/I2 chondrocytes were investigated. The chondrocytes were treated with CTGF or related inhibitors, and transfected with Overexpression and siRNA transfection of Type III Phosphate Transporter 1(Pit-1). Subsequently, the cells were subjected to Alizarin red S staining, PiPer Phosphate Assay Kit, Alkaline Phosphatase Diethanolamine Activity Kit, ELISA, RT-PCR or Western blot analysis. RESULTS Stimulation with Connective tissue growth factor (CTGF) significantly upregulated the expression of the Type III Phosphate Transporter 1(Pit-1) and mineralization levels in chondrocytes through activation of α5β1 integrin and BMP/Samd1/5/8 signaling pathways. Furthermore, treatment with overexpressed Pit-1 markedly increased the expression of Multipass Transmembrane Ankylosis (ANK) transporter in the cells. The inhibitory effect of CTGF receptor blockade using α5β1 Integrin blocking antibody was demonstrated by significantly suppressed the expression of Pit-1 and ANK transporter, as well as chondrocyte mineralization. CONCLUSIONS Our data indicate that Connective tissue growth factor (CTGF) plays a critical role inchondrocyte mineralization, which is dependent on the expression of the Type III Phosphate Transporter 1(Pit-1) and Multipass Transmembrane Ankylosis (ANK) transporter. Consequently, inhibition of CTGF activity may represent a novel therapeutic approach for the management of Osteoarthritis (OA).
Collapse
Affiliation(s)
- Peng Xiao
- Jilin Hospital of Integrated Traditional Chinese and Western Medicine, No. 9, Changchun Road, Jilin, Jilin 132012, PR China.
| | - Yunong Zhu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, PR China.
| | - Hongrui Xu
- Medical College, Dalian University, Dalian, Liaoning 116001, PR China.
| | - Junlei Li
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, PR China.
| | - Angui Tao
- Jilin Hospital of Integrated Traditional Chinese and Western Medicine, Jilin, Jilin 132012, PR China.
| | - Hongji Wang
- Jilin Hospital of Integrated Traditional Chinese and Western Medicine, Jilin, Jilin 132012, PR China.
| | - Dong Cheng
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, PR China.
| | - Xiaojie Dou
- Department of Orthopedics, The First People's Hospital of Huzhou, Huzhou, Zhejiang 313000, PR China.
| | - Lin Guo
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, PR China.
| |
Collapse
|
8
|
Basak S, Hridayanka KSN, Duttaroy AK. Bioactives and their roles in bone metabolism of osteoarthritis: evidence and mechanisms on gut-bone axis. Front Immunol 2024; 14:1323233. [PMID: 38235147 PMCID: PMC10792057 DOI: 10.3389/fimmu.2023.1323233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
Bioactives significantly modify and maintain human health. Available data suggest that Bioactives might play a beneficial role in chronic inflammatory diseases. Although promised, defining their mechanisms and opting to weigh their benefits and limitations is imperative. Detailed mechanisms by which critical Bioactives, including probiotics and prebiotics such as dietary lipids (DHA, EPA, alpha LA), vitamin D, polysaccharides (fructooligosaccharide), polyphenols (curcumin, resveratrol, and capsaicin) potentially modulate inflammation and bone metabolism is limited. Certain dietary bioactive significantly impact the gut microbiota, immune system, and pain response via the gut-immune-bone axis. This narrative review highlights a recent update on mechanistic evidence that bioactive is demonstrated demonstrated to reduce osteoarthritis pathophysiology.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Li YX, Shu J, Kou NN, Chen HB, Guo LM, Yuan Y, He SX, Zhao G. FGF1 reduces cartilage injury in osteoarthritis via regulating AMPK/Nrf2 pathway. J Mol Histol 2023; 54:427-438. [PMID: 37659992 DOI: 10.1007/s10735-023-10143-8] [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: 08/09/2022] [Accepted: 08/07/2023] [Indexed: 09/04/2023]
Abstract
Osteoarthritis (OA) is a systemic joint degenerative disease involving a variety of cytokines and growth factors. In this study, we investigated the protective effect of fibroblast growth factor 1 (FGF1) knockdown on OA and its underlying mechanisms in vitro. In addition, we evaluated the effect of FGF1 knockout on the destabilization of the medial meniscus (DMM) and examined the anterior and posterior cruciate ligament model in vivo. FGF1 affects OA cartilage destruction by increasing the protein expression of Nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1), which is associated with the phosphorylation of AMPK and its substrates. Our study showed that FGF1 knockdown could reverse the oxidative damage associated with osteoarthritis. Nrf2 knockdown eliminated the antioxidant effect of FGF1 knockdown on chondrocytes. Furthermore, AMPK knockdown could stop the impact of FGF1 knockdown on osteoarthritis. These findings suggested that FGF1 knockdown could effectively prevent and reverse osteoarthritis by activating AMPK and Nrf2 in articular chondrocytes.
Collapse
Affiliation(s)
- Yun-Xuan Li
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Jun Shu
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Nan-Nan Kou
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Han-Bo Chen
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Li-Min Guo
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Yong Yuan
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Shao-Xuan He
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China
| | - Gang Zhao
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, 374 Dianmian Avenue, Wuhua District, Kunming, 650000, Yunnan, China.
| |
Collapse
|
10
|
Wang Z, Zhu P, Liao B, You H, Cai Y. Effects and action mechanisms of individual cytokines contained in PRP on osteoarthritis. J Orthop Surg Res 2023; 18:713. [PMID: 37735688 PMCID: PMC10515001 DOI: 10.1186/s13018-023-04119-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Osteoarthritis (OA) is defined as a degenerative joint disease that can affect all tissues of the joint, including the articular cartilage, subchondral bone, ligaments capsule, and synovial membrane. The conventional nonoperative treatments are ineffective for cartilage repair and induce only symptomatic relief. Platelet-rich plasma (PRP) is a platelet concentrate derived from autologous whole blood with a high concentration of platelets, which can exert anti-inflammatory and regenerative effects by releasing multiple growth factors and cytokines. Recent studies have shown that PRP exhibits clinical benefits in patients with OA. However, high operational and equipment requirements greatly limit the application of PRP to OA treatment. Past studies have indicated that high-concentration PRP growth factors and cytokines may be applied as a commercial replacement for PRP. We reviewed the relevant articles to summarize the feasibility and mechanisms of PRP-based growth factors in OA. The available evidence suggests that transforming growth factor-α and β, platelet-derived growth factors, epidermal growth factor, insulin-like growth factor-1, and connective tissue growth factors might benefit OA, while vascular endothelial growth factor, tumor necrosis factor-α, angiopoietin-1, and stromal cell derived factor-1α might induce negative effects on OA. The effects of fibroblast growth factor, hepatocyte growth factor, platelet factor 4, and keratinocyte growth factor on OA remain uncertain. Thus, it can be concluded that not all cytokines released by PRP are beneficial, although the therapeutic action of PRP has a valuable potential to improve.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan, China
| | - Pengfei Zhu
- Department of Cardiovascular, Wuhan Fourth Hospital, Wuhan, China
- Department of Cardiovascular, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bokai Liao
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University and Technology, Jiefang Avenue No.1095, Qiaokou District, Wuhan, 430030, Hubei Province, China.
| | - Yu Cai
- Department of Rehabilitation, Wuhan Fourth Hospital, Hanzheng Street No.473, Qiaokou District, Wuhan, 430000, Hubei Province, China.
| |
Collapse
|
11
|
Hnatjuk MS, Vadzyuk NS, Tatarchuk LV, Monastyrska N. NJ, Konovalenko SO, Yasinovskyi OB. CHANGES IN THE MINERAL COMPOSITION OF THE PATELLA IN DIABETIC KNEE ARTHROPATHY. BULLETIN OF PROBLEMS BIOLOGY AND MEDICINE 2023. [DOI: 10.29254/2077-4214-2022-4-167-285-290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- M. S. Hnatjuk
- I. Horbachevsky Ternopil National Medical University of the Ministry of Health of Ukraine
| | - N. S. Vadzyuk
- I. Horbachevsky Ternopil National Medical University of the Ministry of Health of Ukraine
| | - L. V. Tatarchuk
- I. Horbachevsky Ternopil National Medical University of the Ministry of Health of Ukraine
| | - N. Ja. Monastyrska N.
- I. Horbachevsky Ternopil National Medical University of the Ministry of Health of Ukraine
| | - S. O. Konovalenko
- I. Horbachevsky Ternopil National Medical University of the Ministry of Health of Ukraine
| | - O. B. Yasinovskyi
- I. Horbachevsky Ternopil National Medical University of the Ministry of Health of Ukraine
| |
Collapse
|
12
|
Kawata M, Teramura T, Ordoukhanian P, Head SR, Natarajan P, Sundaresan A, Olmer M, Asahara H, Lotz MK. Krüppel-like factor-4 and Krüppel-like factor-2 are important regulators of joint tissue cells and protect against tissue destruction and inflammation in osteoarthritis. Ann Rheum Dis 2022; 81:1179-1188. [PMID: 35534137 PMCID: PMC9643672 DOI: 10.1136/annrheumdis-2021-221867] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/24/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Analysing expression patterns of Krüppel-like factor (KLF) transcription factors in normal and osteoarthritis (OA) human cartilage, and determining functions and mechanisms of KLF4 and KLF2 in joint homoeostasis and OA pathogenesis. METHODS Experimental approaches included human joint tissues cells, transgenic mice and mouse OA model with viral KLF4 gene delivery to demonstrate therapeutic benefit in structure and pain improvement. Mechanistic studies applied global gene expression analysis and chromatin immunoprecipitation sequencing (ChIP-seq). RESULTS Several KLF genes were significantly decreased in OA cartilage. Among them, KLF4 and KLF2 were strong inducers of cartilage collagen genes and Proteoglycan-4. Cartilage-specific deletion of Klf2 in mature mice aggravated severity of experimental OA. Transduction of human chondrocytes with Adenovirus (Ad) expressing KLF4 or KLF2 enhanced expression of major cartilage extracellular matrix (ECM) genes and SRY-box transcription factor-9, and suppressed mediators of inflammation and ECM-degrading enzymes. Ad-KLF4 and Ad-KLF2 enhanced similar protective functions in meniscus cells and synoviocytes, and promoted chondrocytic differentiation of human mesenchymal stem cells. Viral KLF4 delivery into mouse knees reduced severity of OA-associated changes in cartilage, meniscus and synovium, and improved pain behaviours. ChIP-seq analysis suggested that KLF4 directly bound cartilage signature genes. Ras-related protein-1 signalling was the most enriched pathway in KLF4-transduced cells, and its signalling axis was involved in upregulating cartilage ECM genes by KLF4 and KLF2. CONCLUSIONS KLF4 and KLF2 may be central transcription factors that increase protective and regenerative functions in joint tissue cells, suggesting that KLF gene transfer or molecules upregulating KLFs are therapeutic candidates for OA.
Collapse
Affiliation(s)
- Manabu Kawata
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University, Osaka-Sayama, Osaka, Japan
| | - Philip Ordoukhanian
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Steven R Head
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Padmaja Natarajan
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Aishwarya Sundaresan
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Merissa Olmer
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Hiroshi Asahara
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Martin K Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
13
|
Sheets K, Overbey J, Ksajikian A, Bovid K, Kenter K, Li Y. The pathophysiology and treatment of musculoskeletal fibrosis. J Cell Biochem 2022; 123:843-851. [DOI: 10.1002/jcb.30217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 12/19/2022]
Affiliation(s)
- Kelsey Sheets
- Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine Western Michigan University Kalamazoo Michigan USA
| | - Juliana Overbey
- BioMedical Engineering, Department of Orthopaedic Surgery, WMed, Homer Stryker MD School of Medicine Western Michigan University Kalamazoo Michigan USA
| | - Andre Ksajikian
- BioMedical Engineering, Department of Orthopaedic Surgery, WMed, Homer Stryker MD School of Medicine Western Michigan University Kalamazoo Michigan USA
| | - Karen Bovid
- Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine Western Michigan University Kalamazoo Michigan USA
| | - Keith Kenter
- Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine Western Michigan University Kalamazoo Michigan USA
| | - Yong Li
- Department of Orthopaedic Surgery, Homer Stryker MD School of Medicine Western Michigan University Kalamazoo Michigan USA
| |
Collapse
|
14
|
The essential anti-angiogenic strategies in cartilage engineering and osteoarthritic cartilage repair. Cell Mol Life Sci 2022; 79:71. [PMID: 35029764 PMCID: PMC9805356 DOI: 10.1007/s00018-021-04105-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 01/16/2023]
Abstract
In the cartilage matrix, complex interactions occur between angiogenic and anti-angiogenic components, growth factors, and environmental stressors to maintain a proper cartilage phenotype that allows for effective load bearing and force distribution. However, as seen in both degenerative disease and tissue engineering, cartilage can lose its vascular resistance. This vascularization then leads to matrix breakdown, chondrocyte apoptosis, and ossification. Research has shown that articular cartilage inflammation leads to compromised joint function and decreased clinical potential for regeneration. Unfortunately, few articles comprehensively summarize what we have learned from previous investigations. In this review, we summarize our current understanding of the factors that stabilize chondrocytes to prevent terminal differentiation and applications of these factors to rescue the cartilage phenotype during cartilage engineering and osteoarthritis treatment. Inhibiting vascularization will allow for enhanced phenotypic stability so that we are able to develop more stable implants for cartilage repair and regeneration.
Collapse
|
15
|
Yang Z, Li W, Song C, Leng H. CTGF as a multifunctional molecule for cartilage and a potential drug for osteoarthritis. Front Endocrinol (Lausanne) 2022; 13:1040526. [PMID: 36325449 PMCID: PMC9618584 DOI: 10.3389/fendo.2022.1040526] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2022] Open
Abstract
CTGF is a multifunctional protein and plays different roles in different cells and under different conditions. Pamrevlumab, a monoclonal antibody against CTGF, is an FDA approved drug for idiopathic pulmonary fibrosis (IPF) and Duchenne muscular dystrophy (DMD). Recent studies have shown that CTGF antibodies may potentially serve as a new drug for osteoarthritis (OA). Expression of CTGF is significantly higher in OA joints than in healthy counterparts. Increasing attention has been attracted due to its interesting roles in joint homeostasis. Joint homeostasis relies on normal cellular functions and cell-cell interactions. CTGF is essential for physiological activities of chondrocytes. Abnormal CTGF expression may cause cartilage degeneration. In this review, the physiological functions of CTGF in chondrocytes and related mechanisms are summarized. Changes in the related signaling pathways due to abnormal CTGF are discussed, which are contributing factors to inflammation, cartilage degeneration and synovial fibrosis in OA. The possibility of CTGF as a potential therapeutic target for OA treatment are reviewed.
Collapse
Affiliation(s)
- Zihuan Yang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Chunli Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing Municipal Science & Technology Commission, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- *Correspondence: Huijie Leng,
| |
Collapse
|
16
|
Hnatjuk MS, Vadzyuk NS, Tatarchuk LV, Monastyrska N. NJ, Konovalenko SO, Yasinovskyi OB. MORPHOMETRIC ASPECTS OF STUDY OF FEATURES OF REMODELING OF MICROVESSELS OF SYNOVIAL MEMBRANE OF THE KNEE JOINT AT DIABETIC ARTHROPATHY. BULLETIN OF PROBLEMS BIOLOGY AND MEDICINE 2022. [DOI: 10.29254/2077-4214-2022-3-166-355-363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
17
|
Liu J, Fu Q, Liu S. Transcriptional Regulation Based on Network of Autophagy Identifies Key Genes and Potential Mechanisms in Human Osteoarthritis. Cartilage 2021; 13:1431S-1441S. [PMID: 32819149 PMCID: PMC8804715 DOI: 10.1177/1947603520951632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a chronic arthropathy that frequently occurs in the middle-aged and elderly population. The aim of this study was to investigate the molecular mechanism of OA based on autophagy theory. DESIGN We downloaded the gene expression profile from the Gene Expression Omnibus repository. Differentially expressed genes (DEGs) related to the keyword "autophagy" were identified using the scanGEO online analysis tool. DEGs representing the same expression trend were screened using the MATCH function. Clinical synovial specimens were collected for identification, pathological diagnosis, hematoxylin and eosin staining, and real-time polymerase chain reaction analysis. Differential expression of mRNAs in the synovial membrane tissues and chondrocyte monolayer samples from OA patients was used to identify potential OA biomarkers. Protein-protein interactions were established by the STRING website and visualized with Cytoscape. Functional and pathway enrichment analyses were performed using the Metascape database. RESULTS GABARAPL1, GABARAPL2, and ATG13 were obtained as co-expressed autogenes in the 3 data sets. They were all downregulated among OA synovial tissues compared with non-OA synovial tissues (P < 0.01). A protein-protein interaction network was constructed based on these 3 genes and included 63 genes. A functional analysis revealed that these genes were associated with autophagy-related functions. The top hub genes in the protein-protein interaction network were presented. Furthermore, 3 key modules were extracted to be core control modules. CONCLUSIONS These results offer an important molecular understanding of the key transcriptional regulatory genes and modules based on the network of potential autophagy mechanisms in human OA.
Collapse
Affiliation(s)
- Jiamei Liu
- Department of Pathology, The Shengjing
Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of
China
| | - Qin Fu
- Department of Orthopedics, The Shengjing
Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of
China
| | - Shengye Liu
- Department of Orthopedics, The Shengjing
Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of
China,Shengye Liu, Department of Orthopedics, The
Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004,
People’s Republic of China.
| |
Collapse
|
18
|
Development and Application of 3D Bioprinted Scaffolds Supporting Induced Pluripotent Stem Cells. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4910816. [PMID: 34552987 PMCID: PMC8452409 DOI: 10.1155/2021/4910816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Three-dimensional (3D) bioprinting is a revolutionary technology that replicates 3D functional living tissue scaffolds in vitro by controlling the layer-by-layer deposition of biomaterials and enables highly precise positioning of cells. With the development of this technology, more advanced research on the mechanisms of tissue morphogenesis, clinical drug screening, and organ regeneration may be pursued. Because of their self-renewal characteristics and multidirectional differentiation potential, induced pluripotent stem cells (iPSCs) have outstanding advantages in stem cell research and applications. In this review, we discuss the advantages of different bioinks containing human iPSCs that are fabricated by using 3D bioprinting. In particular, we focus on the ability of these bioinks to support iPSCs and promote their proliferation and differentiation. In addition, we summarize the applications of 3D bioprinting with iPSC-containing bioinks and put forward new views on the current research status.
Collapse
|
19
|
MacDonald IJ, Huang CC, Liu SC, Lin YY, Tang CH. Targeting CCN Proteins in Rheumatoid Arthritis and Osteoarthritis. Int J Mol Sci 2021; 22:ijms22094340. [PMID: 33919365 PMCID: PMC8122640 DOI: 10.3390/ijms22094340] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022] Open
Abstract
The CCN family of matricellular proteins (CYR61/CCN1, CTGF/CCN2, NOV/CCN3 and WISP1-2-3/CCN4-5-6) are essential players in the key pathophysiological processes of angiogenesis, wound healing and inflammation. These proteins are well recognized for their important roles in many cellular processes, including cell proliferation, adhesion, migration and differentiation, as well as the regulation of extracellular matrix differentiation. Substantial evidence implicates four of the proteins (CCN1, CCN2, CCN3 and CCN4) in the inflammatory pathologies of rheumatoid arthritis (RA) and osteoarthritis (OA). A smaller evidence base supports the involvement of CCN5 and CCN6 in the development of these diseases. This review focuses on evidence providing insights into the involvement of the CCN family in RA and OA, as well as the potential of the CCN proteins as therapeutic targets in these diseases.
Collapse
Affiliation(s)
- Iona J. MacDonald
- Graduate Institute of Basic Medical Science, Collage of Medicine, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (Y.-Y.L.)
| | - Chien-Chung Huang
- School of Medicine, Collage of Medicine, China Medical University, Taichung 406040, Taiwan;
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung 404332, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin 65152, Taiwan;
| | - Yen-You Lin
- Graduate Institute of Basic Medical Science, Collage of Medicine, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (Y.-Y.L.)
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, Collage of Medicine, China Medical University, Taichung 40402, Taiwan; (I.J.M.); (Y.-Y.L.)
- School of Medicine, Collage of Medicine, China Medical University, Taichung 406040, Taiwan;
- Graduate Institute of Biomedical Sciences, Collage of Medicine, China Medical University, Taichung 406040, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 406040, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung 413305, Taiwan
- Correspondence:
| |
Collapse
|
20
|
Luo Q, Luo J, Wang Y. YAP Deficiency Attenuates Pulmonary Injury Following Mechanical Ventilation Through the Regulation of M1/M2 Macrophage Polarization. J Inflamm Res 2020; 13:1279-1290. [PMID: 33408500 PMCID: PMC7781043 DOI: 10.2147/jir.s288244] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/15/2020] [Indexed: 12/25/2022] Open
Abstract
Background Evidences indicate that the balance between macrophage M1 and M2 polarization is essential for the regulation of pulmonary inflammation during mechanical ventilation (MV). Yes-associated protein (YAP) is a key component of the Hippo pathway and was suggested to regulate macrophage polarization. This study was designed to investigate whether YAP contributes to pulmonary inflammation during MV. Methods Wild-type and macrophage YAP knockout mice were mechanically ventilated for 12 hours to induce pulmonary injuries. At the end of MV, animals were sacrificed for pulmonary tissue collection and macrophage isolation. In addition, the induction of macrophage polarization was performed in isolated macrophages with or without YAP overexpression in vitro. Pulmonary injuries, YAP expression, macrophage polarization and cytokines were measured. Results Here, we show that MV induces lung injury together with pulmonary inflammation as well as upregulated YAP expressions in pulmonary macrophages. In addition, our results indicate that YAP deficiency in macrophages attenuates pulmonary injury, accompanied with decreased production of pro-inflammatory cytokines including IL (interleukin)-1β, IL-6 and tumor necrosis factor-alpha (TNF-α). Moreover, both in vivo and in vitro studies indicate that YAP deficiency enhances M2 polarization while inhibits M1 polarization. In contrast, YAP overexpression inhibits the induction of M2 polarization but improves M1 polarization. Conclusion Our results report for the first time that the induction of YAP in macrophages contributes to pulmonary inflammation during MV through the regulation of M1/M2 polarization.
Collapse
Affiliation(s)
- Qiong Luo
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China
| | - Jing Luo
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China
| | - Yanlin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, People's Republic of China
| |
Collapse
|
21
|
Chen Z, Zhang N, Chu HY, Yu Y, Zhang ZK, Zhang G, Zhang BT. Connective Tissue Growth Factor: From Molecular Understandings to Drug Discovery. Front Cell Dev Biol 2020; 8:593269. [PMID: 33195264 PMCID: PMC7658337 DOI: 10.3389/fcell.2020.593269] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/09/2020] [Indexed: 01/18/2023] Open
Abstract
Connective tissue growth factor (CTGF) is a key signaling and regulatory molecule involved in different biological processes, such as cell proliferation, angiogenesis, and wound healing, as well as multiple pathologies, such as tumor development and tissue fibrosis. Although the underlying mechanisms of CTGF remain incompletely understood, a commonly accepted theory is that the interactions between different protein domains in CTGF and other various regulatory proteins and ligands contribute to its variety of functions. Here, we highlight the structure of each domain of CTGF and its biology functions in physiological conditions. We further summarized main diseases that are deeply influenced by CTGF domains and the potential targets of these diseases. Finally, we address the advantages and disadvantages of current drugs targeting CTGF and provide the perspective for the drug discovery of the next generation of CTGF inhibitors based on aptamers.
Collapse
Affiliation(s)
- Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
22
|
Shi FL, Ren LX. Up-regulated miR-374a-3p relieves lipopolysaccharides induced injury in CHON-001 cells via regulating Wingless-type MMTV integration site family member 5B. Mol Cell Probes 2020; 51:101541. [PMID: 32092330 DOI: 10.1016/j.mcp.2020.101541] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is a frequent and incurable joint disease, inducing significant pain and seriously threatening to human health. It has been reported that microRNAs (miRNAs) play crucial roles on cancers and inflammatory diseases via cooperating with genes. However, the effect of miR-374a-3p/Wingless-type MMTV integration site family, member 5B (WNT5B) pair in OA remains to be explored. METHODS GSE105027 and GSE55457 datasets were obtained to reveal the expression of miR-374a-3p and WNT5B in OA cartilages using log-scale. The OA cell model was established by lipopolysaccharides (LPS) stimulation in CHON-001 cells and the functional role of miR-374a-3p on OA was investigated by analyzing cell proliferation, cell apoptosis and the expression of apoptosis-related proteins (Bcl-2, Bax and Bim). Through bioinformatics prediction, WNT5B, the target gene of miR-374a-3p, was predicted and the association between miR-374a-3p and WNT5B was further explored by luciferase reporter assay. Functional experiments in vitro were conducted to assess whether WNT5B was involved in the regulation of miR-374a-3p to LPS-stimulated CHON-001. Finally, the expression of JNK/ERK/MAPK pathway-related proteins was detected to explore the underlying molecular mechanism. RESULTS The data set showed that miR-374a-3p was decreased in OA cartilages and the consistent expressional pattern was observed in LPS-stimulated CHON-001 cells. Overexpression of miR-374a-3p significantly alleviated LPS-induced damage in CHON-001 cells, whereas miR-374a-3p inhibitor aggravated LPS-stimulated injury. Further experiments demonstrated that WNT5B was a target of miR-374a-3p and its expression was decreased by miR-374a-3p. WNT5B expression was increased in OA cartilages. Silencing WNT5B prevented CHON-001 cells from LPS-induced damage. Down-regulation of WNT5B strengthened the protective effect of miR-374a-3p on LPS-stimulated CHON-001 cells. Moreover, miR-374a-3p cooperated with WNT5B to affect cell behaviors of LPS-stimulated CHON-001 cells via mediating the JNK/ERK/MAPK pathway. CONCLUSION These results indicated that overexpression of miR-374a-3p protects CHON-001 cells against LPS challenge by modulating WNT5B and inhibiting the JNK/ERK/MAPK pathway.
Collapse
Affiliation(s)
- Feng-Lei Shi
- Department of Orthopaedics, Qilu Hospital of Shandong University, Qingdao, 262021, PR China
| | - Li-Xia Ren
- Department of Rehabilitation, Qilu Hospital of Shandong University, Qingdao, 262021, PR China.
| |
Collapse
|
23
|
Ding LB, Li Y, Liu GY, Li TH, Li F, Guan J, Wang HJ. Long non-coding RNA PVT1, a molecular sponge of miR-26b, is involved in the progression of hyperglycemia-induced collagen degradation in human chondrocytes by targeting CTGF/TGF- β signal ways. Innate Immun 2019; 26:204-214. [PMID: 31625803 PMCID: PMC7144035 DOI: 10.1177/1753425919881778] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The current study was conducted to investigate the role of long non-coding RNA
PVT1 in hyperglycemia-triggered human osteoarthritis (OA) chondrocytes.
Cartilage from knee OA patients with and without diabetes, as well as normal
cartilage, was obtained. Isolated human chondrocytes were treated with 30 nM of
Glc with or without pioglitazone. The expression levels of PVT1, miR-26b, and
type II collagen were determined by RT-PCR. Type II collagen was detected by
immunocytochemistry and chondrocytes were stained with Alcian blue. Moreover,
the interaction among PVT1, miR-26b, and CTGF was examined using bioinformatics,
FISH, RIP, RNA-pull down, and luciferase reporter assays. Over-expression of
PVT1 and miR-26b were performed and expressions of CTGF, TGF-β1, SMAD3, MMP-13,
and type II collagen proteins were examined. Significantly higher expression of
PVT1 was observed in diabetic OA. High Glc induced the elevated expression of
PVT1, CTGF, TGF-β1, IL-6, and MMP-13, as well as decreased expression of type II
collagen and miR-26b. These alterations could be reversed by pioglitazone. PVT1
acted as a sponge for miR-26b to facilitate CTGF expression. Over-expression of
PVT1 increased the expressions of CTGF, TGF-β1, SMAD3, and MMP-13 and decreased
expression of type II collagen. Our findings confirmed that PVT1 is involved in
the hyperglycemia-induced collagen degradation, via the
miR-26b-CTGF-TGF-β1-axis.
Collapse
Affiliation(s)
- Luo-Bin Ding
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, HeBei Province, China
| | - Yao Li
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, HeBei Province, China
| | - Guang-Yuan Liu
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, HeBei Province, China
| | - Tai-Hang Li
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, HeBei Province, China
| | - Feng Li
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, HeBei Province, China
| | - Jian Guan
- Department of Orthopedic Surgery, Third Hospital of Shijiazhuang, Shijiazhuang, HeBei Province, China
| | - Hua-Jun Wang
- Department of Orthopedic Surgery and Sports Medicine Center, First Affiliated Hospital of Jinan University, Guangzhou, GuangDong Province, China
| |
Collapse
|