51
|
Salehi S, Brambilla S, Rasponi M, Lopa S, Moretti M. Development of a Microfluidic Vascularized Osteochondral Model as a Drug Testing Platform for Osteoarthritis. Adv Healthc Mater 2024; 13:e2402350. [PMID: 39370575 DOI: 10.1002/adhm.202402350] [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: 06/28/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by changes in cartilage and subchondral bone. To date, there are no available drugs that can counteract the progression of OA, partly due to the inadequacy of current models to recapitulate the relevant cellular complexity. In this study, an osteochondral microfluidic model is developed using human primary cells to mimic an OA-like microenvironment and this study validates it as a drug testing platform. In the model, the cartilage compartment is created by embedding articular chondrocytes in fibrin hydrogel while the bone compartment is obtained by embedding osteoblasts, osteoclasts, endothelial cells, and mesenchymal stem cells in a fibrin hydrogel enriched with calcium phosphate nanoparticles. After developing and characterizing the model, Interleukin-1β is applied to induce OA-like conditions. Subsequently, the model potential is evaluated as a drug testing platform by assessing the effect of two anti-inflammatory drugs (Interleukin-1 Receptor antagonist and Celecoxib) on the regulation of inflammation- and matrix degradation-related markers. The model responded to inflammation and demonstrated differences in drug efficacy. Finally, it compares the behavior of the "Cartilage" and "Cartilage+Bone" models, emphasizing the necessity of incorporating both cartilage and bone compartments to capture the complex pathophysiology of OA.
Collapse
Affiliation(s)
- Shima Salehi
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Stefania Brambilla
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, Milan, 20157, Italy
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Via Chiesa 5, Bellinzona, 6500, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Via Tesserete 46, Lugano, 6900, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Via Buffi 13, Lugano, 6900, Switzerland
| |
Collapse
|
52
|
Giacomini F, Rho HS, Eischen‐Loges M, Tahmasebi Birgani Z, van Blitterswijk C, van Griensven M, Giselbrecht S, Habibović P, Truckenmüller R. Enthesitis on Chip - A Model for Studying Acute and Chronic Inflammation of the Enthesis and its Pharmacological Treatment. Adv Healthc Mater 2024; 13:e2401815. [PMID: 39188199 PMCID: PMC11650547 DOI: 10.1002/adhm.202401815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Enthesitis, the inflammation of the enthesis, which is the point of attachment of tendons and ligaments to bones, is a common musculoskeletal disease. The inflammation often originates from the fibrocartilage region of the enthesis as a consequence of mechanical overuse or -load and consequently tissue damage. During enthesitis, waves of inflammatory cytokines propagate in(to) the fibrocartilage, resulting in detrimental, heterotopic bone formation. Understanding of human enthesitis and its treatment options is limited, also because of lacking in vitro model systems that can closely mimic the pathophysiology of the enthesis and can be used to develop therapies. In this study, an enthes(it)is-on-chip model is developed. On opposite sides of a porous culture membrane separating the chip's two microfluidic compartments, human mesenchymal stromal cells are selectively differentiated into tenocytes and fibrochondrocytes. By introducing an inflammatory cytokine cocktail into the fibrochondrocyte compartment, key aspects of acute and chronic enthesitis, measured as increased expression of inflammatory markers, can be recapitulated. Upon inducing chronic inflammatory conditions, hydroxyapatite deposition, enhanced osteogenic marker expression and reduced secretion of tissue-related extracellular matrix components are observed. Adding the anti-inflammatory drug celecoxib to the fibrochondrocyte compartment mitigates the inflammatory state, demonstrating the potential of the enthesitis-on-chip model for drug testing.
Collapse
Affiliation(s)
- Francesca Giacomini
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Hoon Suk Rho
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Maria Eischen‐Loges
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Martijn van Griensven
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|
53
|
Jeon Y, Kim J, Kwon H, Yeon YJ, Kim T, Ham J, Kim YJ. Cannabiorcol as a novel inhibitor of the p38/MSK-1/NF-κB signaling pathway, reducing matrix metalloproteinases in osteoarthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156141. [PMID: 39405610 DOI: 10.1016/j.phymed.2024.156141] [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: 05/28/2024] [Revised: 09/21/2024] [Accepted: 10/07/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND The bioactivity and potential medicinal applications of cannabiorcol, a lesser-known derivative of Cannabis sativa, require further investigation. Osteoarthritis (OA) is a chronic joint condition marked by gradual degradation of the cartilage and commonly associated with elevated levels of matrix metalloproteinases (MMPs). However, the influence of cannabiorcol on OA and its underlying mechanisms remains unclear. METHODS In silico analysis investigated the key transcription factors that regulate MMP expression. A chondrocyte cell model [interleukin (IL)-1β and IL-1⍺-treated C20A4 cell line] was established and treated with cannabiorcol. Associated cytotoxicity was assessed using a WST-8 assay. A monoiodoacetate-induced OA rat model was established and treated with cannabiorcol. Protein translocation and transactivation analyses were conducted using immunofluorescence and dual-luciferase reporter assays, respectively. Western blotting and real-time PCR analyzed relevant markers to examine cannabiorcol's effects on OA and its fundamental mechanisms. RESULTS Cannabiorcol inhibits the expression of IL-1β-induced MMPs compared to other cannabis-related compounds. In silico analysis revealed that the nuclear factor-kappa β (NF-κβ) and mitogen-activated protein kinase (MAPK) pathways are associated with MMP expression as key regulators. In vitro, cannabiorcol inhibits the NF-κB and p38 MAPK pathways independently cannabinoid receptors and transient receptor potential vanilloids. In vivo, cannabiorcol reduces MMP expression and ameliorates monoiodoacetate-induced OA traits in rats. CONCLUSION Cannabiorcol inhibits IL-1β-induced MMP expression in vitro and alleviates OA in an MIA-induced OA rat model by reducing MMP expression and inhibiting the p65/p38 axis.
Collapse
Affiliation(s)
- Youngsic Jeon
- Institute of Natural Products, Korea Institute of Science and Technology, Republic of Korea
| | - Jiyool Kim
- Institute of Natural Products, Korea Institute of Science and Technology, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Republic of Korea
| | - Hyukjoon Kwon
- Institute of Natural Products, Korea Institute of Science and Technology, Republic of Korea
| | - Young Joo Yeon
- Department of Biochemical Engineering, Gangneung-Wonju National University, Republic of Korea
| | - Taejung Kim
- Institute of Natural Products, Korea Institute of Science and Technology, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Republic of Korea.
| | - Jungyeob Ham
- Institute of Natural Products, Korea Institute of Science and Technology, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Republic of Korea; NeoCannBio Co., Ltd., Republic of Korea.
| | - Young-Joo Kim
- Institute of Natural Products, Korea Institute of Science and Technology, Republic of Korea.
| |
Collapse
|
54
|
Kwon EJ, Lee H, Shin U, Kim ES, Myung K, Kim J, Park JH, Kim K, Lee Y, Oh CK, Kim YH. Ionizing radiation inhibits zebrafish embryo hatching through induction of tissue inhibitors of metalloproteinases (TIMPs) expression. FEBS J 2024; 291:5470-5485. [PMID: 39547957 DOI: 10.1111/febs.17318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Ionizing radiation (IR) has garnered growing attention because of its biological effects on aquatic organisms and humans. Here, we identify the most impacted organs and uncover the molecular mechanisms causing the changes in the context of vertebrate development using single-cell RNA sequencing. Alterations in cellular composition and biological functions were explored using transcriptomic profiling of zebrafish embryos exposed to 5 Gy. Single-cell RNA sequencing analyses unveiled notable shifts in the proportions of brain/central nervous system and hatching gland clusters. Although IR exposure led to increased expression of hatching enzymes, a significant but mild delay in hatching was observed following 5 Gy IR exposure. Gene Ontology analysis showed an increased expression of tissue inhibitors of metalloproteinases (TIMPs), known as matrix metalloproteinase inhibitors, which was confirmed via whole-mount in situ hybridization. Correlation analysis linked TIMPs to transcription factors cebpb and cebpd, which were significantly correlated post-IR exposure. Although no morphological changes were observed in some organs, including the brain, the study reveals substantial alterations in developing vertebrates. Notably, despite increased hatching enzymes, elevated TIMPs in the hatching gland suggest a regulatory mechanism impacting hatching activity. This research contributes to comprehending the ecological repercussions of IR exposure, emphasizing the importance of safety measures for aquatic ecosystems and overall environmental health.
Collapse
Affiliation(s)
- Eun Jung Kwon
- Medical Research Institute, Pusan National University, Yangsan, Korea
| | - Hansong Lee
- Medical Research Institute, Pusan National University, Yangsan, Korea
| | - Unbum Shin
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Eun-Sun Kim
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Jeongmo Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Jung-Hoon Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Kihun Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yoonsung Lee
- Research Institute of Clinical Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Kyu Oh
- Department of Biochemistry, School of Medicine, Pusan National University, Yangsan, Korea
- Institute for Future Earth, Pusan National University, Busan, Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
55
|
Morcos CA, Haiba NS, Bassily RW, Abu-Serie MM, El-Yazbi AF, Soliman OA, Khattab SN, Teleb M. Structure optimization and molecular dynamics studies of new tumor-selective s-triazines targeting DNA and MMP-10/13 for halting colorectal and secondary liver cancers. J Enzyme Inhib Med Chem 2024; 39:2423174. [PMID: 39513468 PMCID: PMC11552285 DOI: 10.1080/14756366.2024.2423174] [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: 10/27/2023] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
A series of triazole-tethered triazines bearing pharmacophoric features of DNA-targeting agents and non-hydroxamate MMPs inhibitors were synthesized and screened against HCT-116, Caco-2 cells, and normal colonocytes by MTT assay. 7a and 7g surpassed doxorubicin against HCT-116 cells regarding potency (IC50 = 0.87 and 1.41 nM) and safety (SI = 181.93 and 54.41). 7g was potent against liver cancer (HepG-2; IC50 = 65.08 nM), the main metastatic site of CRC with correlation to MMP-13 expression. Both derivatives induced DNA damage at 2.67 and 1.87 nM, disrupted HCT-116 cell cycle and triggered apoptosis by 33.17% compared to doxorubicin (DNA damage at 0.76 nM and 40.21% apoptosis induction). 7g surpassed NNGH against MMP-10 (IC50 = 0.205 μM) and MMP-13 (IC50 = 0.275 μM) and downregulated HCT-116 VEGF related to CRC progression by 38%. Docking and MDs simulated ligands-receptors binding modes and highlighted SAR. Their ADMET profiles, drug-likeness and possible off-targets were computationally predicted.
Collapse
Affiliation(s)
- Christine A. Morcos
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Nesreen S. Haiba
- Department of Physics and Chemistry, Faculty of Education, Alexandria University, Alexandria, Egypt
| | - Rafik W. Bassily
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Marwa M. Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Egypt
| | - Amira F. El-Yazbi
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Omar A. Soliman
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
| |
Collapse
|
56
|
Shi H, Wang H, Yu M, Su J, Zhao Z, Gao T, Zhang Q, Wei Y. Serum trace elements and osteoarthritis: A meta-analysis and Mendelian randomization study. J Trace Elem Med Biol 2024; 86:127520. [PMID: 39255532 DOI: 10.1016/j.jtemb.2024.127520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/23/2024] [Accepted: 09/01/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE This study aims to establish the correlation between shifts in serum trace element (TE) levels and the progression of osteoarthritis (OA), while also exploring the underlying causal relationship between these variables. METHODS An investigation was conducted, which included a systematic review, a meta-analysis of observational studies, and a two-sample Mendelian randomization (MR) study. RESULTS This meta-analysis revealed significant differences in serum levels of copper, manganese, cadmium, and selenium between OA patients and healthy controls, after adjusting for heterogeneity. Specifically, significant disparities were observed for copper (SMD 0.118 [95 % CI: 0.061 ∼ 0.175], P < 0.001), manganese (SMD -0.180 [95 % CI: -0.326 ∼ -0.034], P = 0.016), cadmium (SMD 0.227 [95 % CI: 0.131 ∼ 0.322], P < 0.001), and selenium (SMD -0.138 [95 % CI: -0.209 ∼ -0.068], P < 0.001), while zinc levels did not show a significant difference (SMD -0.02 [95 % CI: -0.077 ∼ 0.038], P = 0.503). Further, MR analysis suggested a causal link between genetically predicted serum copper level changes and OA development, but not for other TEs. CONCLUSION The study suggests that there is an association between the occurrence of OA and variations in serum levels of copper, manganese, cadmium, and selenium. Elevated serum copper may play a pivotal role. Further research is needed to explore the therapeutic potential of TE level modulation in OA management.
Collapse
Affiliation(s)
- Haoyan Shi
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haochen Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minghao Yu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianbang Su
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ze Zhao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianqi Gao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qian Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingliang Wei
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
57
|
Tai C, Ito A, Zhao Z, Kuroki H, Aoyama T. Attenuating Cartilage Degeneration in a Low Mechanical Compression Rat Model Through Intra-Articular Injections of Allogeneic Bone Marrow-Derived Mesenchymal Stem Cells. Cartilage 2024:19476035241301291. [PMID: 39611391 DOI: 10.1177/19476035241301291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVE Mechanical stimulation significantly contributes to posttraumatic osteoarthritis (PTOA), a condition that impedes patient recovery following intra-articular injury. Effective treatment options for compression-induced injuries are limited. Bone marrow-derived mesenchymal stem cell (BMSC) implantation has emerged as a potential therapeutic breakthrough for joint diseases. The aim of this study was to attenuate the progression of PTOA induced by cyclic loading and demonstrate the potential effectiveness of BMSCs in a rat model of low mechanical compression. DESIGN Using a rat model of compression-induced articular cartilage injury, assessments were conducted 2, 4, and 8 weeks after cyclic compressive loading. The expression of matrix metallopeptidase 13, transforming growth factor-beta 3 (TGF-β3), insulin-like growth factor 1 (IGF-1), and cleaved caspase-3 was evaluated through immunohistochemistry to investigate the mechanistic aspects underlying the prevention of compression-induced injury following BMSCs treatment. RESULTS Intra-articular injections of BMSCs significantly improved scores in the OARSI (Osteoarthritis Research Society International) Osteoarthritis Cartilage Histopathology Assessment System and Histological-Histochemical Grading System. This treatment showed positive outcomes in maintaining high relative cell density and reducing proteoglycan loss after cyclic compression-induced injury. The expression patterns of IGF-1 and TGF-β3 provide valuable insights into the presence and distribution of these growth factors in healthy and injured cartilage. CONCLUSIONS These findings highlight the efficacy of BMSCs treatment in attenuating the advancement of compression-induced injuries, albeit within a limited timeframe.
Collapse
Affiliation(s)
- Chia Tai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Zixi Zhao
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kuroki
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
58
|
Song JH, Koh H, Moon HY, Jung JG, Hong JH, Sunwoo J. Comparison of a fixed-dose combination of Celecoxib/PG201 [Layla®] versus co-administration of individual formulations in healthy participants: A randomized trial. Medicine (Baltimore) 2024; 103:e40494. [PMID: 39560548 PMCID: PMC11575945 DOI: 10.1097/md.0000000000040494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent joint disease affecting the spine, hands, hips, knees, and feet. However, definitive drugs for OA are lacking, and current treatments are limited owing to inconvenient administration, inadequate functional improvement, and long-term side effects including gastrointestinal and cardiovascular adverse events. Therefore, in this study, we aimed to assess the pharmacokinetics and safety profiles of PK101, a fixed-dose combination (FDC) comprising PG201, a 12-herb extract used in OA treatment in traditional East Asian medicine, and celecoxib, a selective cyclooxygenase-2 inhibitor, by comparing its administration as an FDC and the corresponding individual formulations in healthy subjects. PATIENTS AND METHODS A randomized, open-label, single-dose, 2 × 2 crossover design with a cohort of healthy participants. All subjects received a single FDC tablet (405.4 mg PG201 and 100 mg celecoxib) or the individual formulations, with 7-day washout period between administrations. The estimation of maximum plasma concentration and area under the plasma concentration-time curve from time zero to the time of the last quantifiable concentration of celecoxib involved determining the geometric mean ratios and 90% confidence intervals of the FDC compared to its individual formulations. RESULTS Forty-six participants were enrolled; however, only 44 completed the study. The geometric mean ratios (90% confidence intervals) for the area under the plasma concentration-time curve from time zero to the time of the last quantifiable concentration and maximum plasma concentration of celecoxib were 1.1124 (1.0601-1.1672) and 1.2788 (1.1708-1.3969), respectively. The time of maximum plasma concentration range was 1.0 to 4.0 hours and 1.0 to 6.0 hours (minimum-maximum) for the FDC and individual formulations, respectively. Seven adverse events occurred in 6 subjects. CONCLUSION The systemic exposure and safety profiles of the individual and FDC formulations were similar, supporting their potential as an innovative and effective therapeutic approach for OA treatment. All relevant data are within the paper and its Supporting Information files.
Collapse
Affiliation(s)
- Ji Hye Song
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Hyunsook Koh
- PMG Pharm Co., Ltd., Gyeonggi-do, Republic of Korea
| | | | - Jin-Gyu Jung
- Department of Family Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jang Hee Hong
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Jung Sunwoo
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Republic of Korea
| |
Collapse
|
59
|
Soh LJ, Lee SY, Roebuck MM, Wong PF. Unravelling the interplay between ER stress, UPR and the cGAS-STING pathway: Implications for osteoarthritis pathogenesis and treatment strategy. Life Sci 2024; 357:123112. [PMID: 39378929 DOI: 10.1016/j.lfs.2024.123112] [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: 04/29/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/10/2024]
Abstract
Osteoarthritis (OA) is a debilitating chronic degenerative disease affecting the whole joint organ leading to pain and disability. Cellular stress and injuries trigger inflammation and the onset of pathophysiological changes ensue after irreparable damage and inability to resolve inflammation, impeding the completion of the healing process. Extracellular matrix (ECM) degradation leads to dysregulated joint tissue metabolism. The reparative effort induces the proliferation of hypertrophic chondrocytes and matrix protein synthesis. Aberrant protein synthesis leads to endoplasmic reticulum (ER) stress and chondrocyte apoptosis with consequent cartilage matrix loss. These events in a vicious cycle perpetuate inflammation, hindering the restoration of normal tissue homeostasis. Recent evidence suggests that inflammatory responses and chondrocyte apoptosis could be caused by the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signalling axis in response to DNA damage. It has been reported that there is a crosstalk between ER stress and cGAS-STING signalling in cellular senescence and other diseases. Based on recent evidence, this review discusses the role of ER stress, Unfolded Protein Response (UPR) and cGAS-STING pathway in mediating inflammatory responses in OA.
Collapse
Affiliation(s)
- Li-Jen Soh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Siam-Yee Lee
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Margaret M Roebuck
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool L3 9TA, UK
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia.
| |
Collapse
|
60
|
Byun S, Coryell P, Kramer N, D’Costa S, Thulson E, Shine J, Parkus S, Chubinskaya S, Loeser RF, Diekman BO, Phanstiel DH. Response splicing QTLs in primary human chondrocytes identifies putative osteoarthritis risk genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622754. [PMID: 39605710 PMCID: PMC11601258 DOI: 10.1101/2024.11.11.622754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Osteoarthritis affects millions worldwide, yet effective treatments remain elusive due to poorly understood molecular mechanisms. While genome-wide association studies (GWAS) have identified over 100 OA-associated loci, identifying the genes impacted at each locus remains challenging. Several studies have mapped expression quantitative trait loci (eQTL) in chondrocytes and colocalized them with OA GWAS variants to identify putative OA risk genes; however, the degree to which genetic variants influence OA risk via alternative splicing has not been explored. We investigated the role of alternative splicing in OA pathogenesis using RNA-seq data from 101 human chondrocyte samples treated with PBS (control) or fibronectin fragment (FN-f), an OA trigger. We identified 590 differentially spliced genes between conditions, with FN-f inducing splicing events similar to those in primary OA tissue. We used CRISPR/Cas9 to mimic an SNRNP70 splicing event observed in OA and FN-f-treated chondrocytes and found that it induced an OA-like expression pattern. Integration with genotyping data revealed 7,188 splicing quantitative trait loci (sQTL) affecting 3,056 genes. While many sQTLs were shared, we identified 738 and 343 condition-specific sQTLs for control and FN-f, respectively. We identified 15 RNA binding proteins whose binding sites were enriched at sQTL splice junctions and found that expression of those RNA binding proteins correlated with exon inclusion. Colocalization with OA GWAS identified 6 putative risk genes, including a novel candidate, PBRM1. Our study highlights the significant impact of alternative splicing in OA and provides potential therapeutic targets for future research.
Collapse
Affiliation(s)
- Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nicole Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D’Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan Chubinskaya
- Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Dvision of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
61
|
Qiao L, Li Z, Li B, Zhang F, Yao Z, Wu C, Tang H, Pan Q, Shi P, Ping Y. Combination of anti-inflammatory therapy and RNA interference by light-inducible hybrid nanomedicine for osteoarthritis treatment. Acta Pharm Sin B 2024; 14:5008-5025. [PMID: 39664429 PMCID: PMC11628851 DOI: 10.1016/j.apsb.2024.06.009] [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: 04/03/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 12/13/2024] Open
Abstract
Osteoarthritis (OA) is a type of highly prevalent heterogeneous degenerative disease that leads to joint pain, deformity, the destruction of articular cartilage, and eventual disability. The current treatment strategies for OA often suffer from systemic side effects, poor anti-inflammatory efficacy, and persistent pain. To address these issues, we develop light-inducible nanomedicine that enables the co-delivery of anti-inflammatory drug (diacerein, DIA) and small interfering RNA (siRNA) targeting nerve growth factor (NGF) for pain relief to enhance the therapeutic efficacy of OA. The nanomedicine is based on poly(β-amino-ester)-coated gold nanocages (AuNCs), which is further incorporated with the phase-change material (lauric acid/stearic acid, LA/SA). Following intra-articular (IA) injection in vivo, the nanomedicine displays high degree of drug accumulation and retention in the joint lesion of OA mouse models. The photothermal effect, induced by AuNCs, not only promotes DIA and siRNA release, but also upregulates the expression of heat shock protein 70 (HSP-70) to resist the apoptosis of chondrocytes in the inflammatory condition. The internalization of both DIA and siRNA results in strong anti-inflammatory and pain-relieving effects, which greatly contribute to the joint repair of OA mice. This study offers a promising combination strategy for OA treatment.
Collapse
Affiliation(s)
- Li Qiao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Zhiyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bowen Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhuo Yao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chongzhi Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honglin Tang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
| | - Qi Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Shi
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou 310016, China
| | - Yuan Ping
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou 310016, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| |
Collapse
|
62
|
Wu Y, Gong Y, Liu L, Bai L, Zhang Y, Li S, Wang C, Yuan Y, Lv X, Qin Y, Wang H, Liu Y, Chen F, Chen S, Zhang F, Guo X, Wang X, Ning Y. The Impact of Selenium Deficiency and T-2 Toxin on Zip6 Expression in Kashin-Beck Disease. Biol Trace Elem Res 2024:10.1007/s12011-024-04426-8. [PMID: 39455492 DOI: 10.1007/s12011-024-04426-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
This study investigated the expression of Zip6, a gene predominantly located in the placenta, breast, and prostate tissues, in patients with Kashin-Beck disease (KBD). Environmental risk factor models for KBD were developed using low selenium (Se) feeding (with a Se content of 0.02 mg Se/kg in the feed) and exposure to T-2 toxin (200 ng/g*BW/D). Additionally, the study examined the alterations in Se and Zn2+ levels, along with the mRNA and protein expression levels of Zip6 and KBD related genes, including Mtf1, Mmp3, Mmp13, Adamts5, and Col2a1. Differentially expressed genes (DEGs) were examined by transcriptome sequencing to elucidate the mechanism by which Zip6 induces metabolic disorder of the extracellular matrix (ECM), subsequently leading to cartilage injury under the influence of Se deficiency and T-2 toxin. The findings indicated that the expression levels of Zip6 in adult and pediatric KBD chondrocytes were not synchronized. In the animal study, there was a notable increase in the Zn2+ level in the comprehensive exposure (CE) group. Moreover, in both the T-2 exposure (T-2) and CE groups, there was a significant decrease in the expression of Zip6 in each zone, and the expression of Adamts5 in the middle zone exhibited a significant increase (P < 0.05) correlating with varying degrees of cartilage tissue damage in each group. Sequencing results revealed that the significantly up-regulated DEGs in the CE group included Zimz2. This study suggested that Se and T-2 toxin may influence the expression of Zip6, and it investigated the role of Zn2+ in the pathogenesis of KBD, thereby providing a novel scientific foundation for understanding the pathogenesis of KBD.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yi Gong
- Center for Immunological and Metabolic Diseases, MED-X Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Lian Liu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Lulu Bai
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Shujin Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Chaowei Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yuequan Yuan
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xi Lv
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yirong Qin
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Hui Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yanli Liu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Feihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Sijie Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Feiyu Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xiong Guo
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China
- Clinical Research Center for Endemic Disease of Shaanxi Province, the Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, No.157 Xi Wu Road, Xi'an, 710004, People's Republic of China
| | - Xi Wang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
| | - Yujie Ning
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
63
|
He F, Wu H, He B, Han Z, Chen J, Huang L. Antioxidant hydrogels for the treatment of osteoarthritis: mechanisms and recent advances. Front Pharmacol 2024; 15:1488036. [PMID: 39525636 PMCID: PMC11543442 DOI: 10.3389/fphar.2024.1488036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Articular cartilage has limited self-healing ability, resulting in injuries often evolving into osteoarthritis (OA), which poses a significant challenge in the medical field. Although some treatments exist to reduce pain and damage, there is a lack of effective means to promote cartilage regeneration. Reactive Oxygen Species (ROS) have been found to increase significantly in the OA micro-environment. They play a key role in biological systems by participating in cell signaling and maintaining cellular homeostasis. Abnormal ROS expression, caused by internal and external stimuli and tissue damage, leads to elevated levels of oxidative stress, inflammatory responses, cell damage, and impaired tissue repair. To prevent excessive ROS accumulation at injury sites, biological materials can be engineered to respond to the damaged microenvironment, release active components in an orderly manner, regulate ROS levels, reduce oxidative stress, and promote tissue regeneration. Hydrogels have garnered significant attention due to their excellent biocompatibility, tunable physicochemical properties, and drug delivery capabilities. Numerous antioxidant hydrogels have been developed and proven effective in alleviating oxidative stress. This paper discusses a comprehensive treatment strategy that combines antioxidant hydrogels with existing treatments for OA and explores the potential applications of antioxidant hydrogels in cartilage tissue engineering.
Collapse
Affiliation(s)
- Feng He
- Department of Orthopedics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China
| | - Hongwei Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China
| | - Bin He
- Department of Orthopedics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China
| | - Zun Han
- Department of Orthopedics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China
| | - Jiayi Chen
- Department of Orthopedics, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China
| | - Lei Huang
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
64
|
Cesarone MR, Belcaro G, Cox D, Scipione V, Scipione C, Dugall M, Hosoi M, Feragalli B, Hu S, Coppazuccari F, Cotellese R. Supplementary management of symptomatic hand osteoarthritis with Pycnogenol®. Minerva Surg 2024; 79:539-544. [PMID: 38980278 DOI: 10.23736/s2724-5691.24.10403-0] [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: 07/10/2024]
Abstract
BACKGROUND The aim of this 4-week pilot registry, supplement study was to assess the effects of Pycnogenol® compared to a standard management on hand osteoarthritis associated with pain. As Pycnogenol® decreases inflammation and pain, chronic use of drugs, causing side effects may be reduced. METHODS The registry patients included suffered finger pain associated with hand osteoarthritis All subjects used a standard management (SM). A supplementary group additionally used 150 mg Pycnogenol® per day. In addition, a retrospective group with 40 comparable subjects using oral diclofenac was used for comparison. Forty-two subjects with hand osteoarthritis completed the study. The registry patients were former sport professionals, fishermen and subjects working with their hands in a common manual activity. 22 subjects took Pycnogenol® in addition to standard management and 20 subjects followed the standard management only and served as controls. RESULTS The two groups were comparable at inclusion. No subject had to stop supplementation or the SM. No side effects were observed. After 4 weeks, spontaneous pain in the morning and pain after work were significantly reduced with Pycnogenol® supplementation compared to controls (P<0.05). Residual pain at rest in the evening was significantly improved after 4 weeks with the supplement compared to controls (P<0.05). The number of subjects requiring pain medication during the 4-week study period was significantly lower in the supplement group (2/22) compared to controls (8/20) (P<0.05). Hand dynamometry results show significant improvement in hand-finger strength (due to decreased pain and stiffness) with the supplement compared to controls (P<0.05). At inclusion, all subjects presented hyperthermic joints, 2°C higher than the surrounding tissues as shown by thermography. After 4 weeks, the number of subjects with hyperthermic joints was lower in the Pycnogenol® group than in controls (P<0.05). Both nonspecific markers of inflammation (ESR and C-reactive protein levels in blood) were significantly lower after 4 weeks in the Pycnogenol® group than in controls (P<0.05). Other routine blood tests were normal at inclusion and at the end of the study. Within 4 weeks, plasma oxidative stress decreased by 14.4% (P<0.05) in the Pycnogenol® group vs. 5.5% in the control group. The retrospective comparison with a group of 40 comparable subjects using oral diclofenac showed that after 4 weeks, the efficacy of Pycnogenol® on improving pain in the morning, after work and in the evening, on hand-finger strength and on decreasing C-reactive protein was significantly higher (P<0.05) than in the diclofenac group (comparable, non-parallel group, CNPG). CONCLUSIONS In conclusion, supplementation with Pycnogenol® was well tolerated and effectively controlled pain while improving grip strength in patients with hand osteoarthritis. All supplement subjects showed an improved operativity.
Collapse
Affiliation(s)
- Maria R Cesarone
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | - Gianni Belcaro
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy -
| | - David Cox
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | - Valeria Scipione
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | - Claudia Scipione
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | - Mark Dugall
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | - Morio Hosoi
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | - Beatrice Feragalli
- Department of Medical and Oral Sciences and Biotechnologies, D'Annunzio University, Pescara, Italy
| | - Shu Hu
- Irvine3 Labs and PAP/PEA Screening Project, San Valentino, Pescara, Italy
| | | | - Roberto Cotellese
- Department of Medical and Oral Sciences and Biotechnologies, D'Annunzio University, Pescara, Italy
| |
Collapse
|
65
|
Du C, Chen Z, Liu S, Liu J, Zhan J, Zou J, Liao J, Huang W, Lei Y. Lubricin-Inspired Nanozymes Reconstruct Cartilage Lubrication System with an "In-Out" Strategy. SMALL METHODS 2024; 8:e2400757. [PMID: 38962862 DOI: 10.1002/smtd.202400757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Lubricin, secreted primarily by chondrocytes, plays a critical role in maintaining the function of the cartilage lubrication system. However, both external factors such as friction and internal factors like oxidative stress can disrupt this system, leading to osteoarthritis. Inspired by lubricin, a lubricating nanozyme, that is, Poly-2-acrylamide-2-methylpropanesulfonic acid sodium salt-grafted aminofullerene, is developed to restore the cartilage lubrication system using an "In-Out" strategy. The "Out" aspect involves reducing friction through a combination of hydration lubrication and ball-bearing lubrication. Simultaneously, the "In" aspect aims to mitigate oxidative stress by reducing free radical, increasing autophagy, and improving the mitochondrial respiratory chain. This results in reduced chondrocyte senescence and increased lubricin production, enhancing the natural lubrication ability of cartilage. Transcriptome sequencing and Western blot results demonstrate that it enhances the functionality of mitochondrial respiratory chain complexes I, III, and V, thereby improving mitochondrial function in chondrocytes. In vitro and in vivo experiments show that the lubricating nanozymes reduce cartilage wear, improve chondrocyte senescence, and mitigate oxidative stress damage, thereby mitigating the progression of osteoarthritis. These findings provide novel insights into treating diseases associated with oxidative stress and frictional damage, such as osteoarthritis, and set the stage for future research and development of therapeutic interventions.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingdi Zhan
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jing Zou
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junyi Liao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
66
|
Christopoulou ME, Aletras AJ, Papakonstantinou E, Stolz D, Skandalis SS. WISP1 and Macrophage Migration Inhibitory Factor in Respiratory Inflammation: Novel Insights and Therapeutic Potentials for Asthma and COPD. Int J Mol Sci 2024; 25:10049. [PMID: 39337534 PMCID: PMC11432718 DOI: 10.3390/ijms251810049] [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: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, β-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
67
|
Lu C, Yang W, Chu F, Wang S, Ji Y, Liu Z, Yu H, Qin S, Sun D, Jiao Z, Sun H. Hesperetin Attenuates T-2 Toxin-Induced Chondrocyte Injury by Inhibiting the p38 MAPK Signaling Pathway. Nutrients 2024; 16:3107. [PMID: 39339707 PMCID: PMC11434908 DOI: 10.3390/nu16183107] [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: 07/07/2024] [Revised: 09/01/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Hesperetin, a flavonoid derived from citrus fruits, exhibits potent antioxidant and anti-inflammatory activities and has been implicated in cartilage protection. However, its effectiveness against T-2 toxin-induced knee cartilage damage remains unclear. METHODS In this study, high-throughput sequencing analysis was employed to identify the key signaling pathways involved in T-2 toxin-induced articular cartilage damage in rats. Animal models were divided into the following groups: control, low-dose T-2 toxin, high-dose T-2 toxin, T-2 toxin + hesperetin, hesperetin, and vehicle. Pathological staining and immunohistochemistry were used to assess pathological changes, as well as the expression levels of the cartilage matrix-related proteins MMP13 and collagen II, along with the activation of the p38 MAPK signaling pathway. Additionally, primary rat chondrocytes were cultured to establish an in vitro model for investigating the underlying mechanism. RESULTS High-throughput sequencing analysis revealed the involvement of the MAPK signaling pathway in T-2 toxin-induced articular cartilage damage in rats. Hesperetin intervention in T-2 toxin-exposed rats attenuated pathological cartilage damage. Immunohistochemistry results demonstrated a significant reduction in collagen II protein expression in the high-dose T-2 toxin group (p < 0.01), accompanied by a significant increase in MMP13 protein expression (p < 0.01). In both the articular cartilage and the epiphyseal plate, the T-2 toxin + hesperetin group exhibited significantly higher collagen II protein expression than the high-dose T-2 toxin group (p < 0.05), along with significantly lower MMP13 protein expression (p < 0.05). Hesperetin inhibited the over-activation of the p38/MEF2C signaling axis induced by T-2 toxin in primary rat chondrocytes. Compared to the T-2 toxin group, the T-2 toxin + hesperetin group showed significantly reduced phosphorylation levels of p38 and protein expression levels of MEF2C (p < 0.001 or p < 0.05). Moreover, the T-2 toxin + hesperetin group exhibited a significant decrease in MMP13 protein expression (p < 0.05) and a significant increase in collagen II protein expression (p < 0.01) compared to the T-2 toxin group. CONCLUSIONS T-2 toxin activates the p38 MAPK signaling pathway, causing knee cartilage damage in rats. Treatment with hesperetin inhibits the p38/MEF2C signaling axis, regulates collagen II and MMP13 protein expression, and reduces cartilage injury significantly.
Collapse
Affiliation(s)
- Chunqing Lu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Wenjing Yang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Fang Chu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Sheng Wang
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Yi Ji
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
- Institute of Keshan Disease, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Zhipeng Liu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Hao Yu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Shaoxiao Qin
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Dianjun Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| | - Zhe Jiao
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
- Institute for Kashin Beck Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Hongna Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, National Health Commission Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health & Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
68
|
Guan M, Yu Q, Zhou G, Wang Y, Yu J, Yang W, Li Z. Mechanisms of chondrocyte cell death in osteoarthritis: implications for disease progression and treatment. J Orthop Surg Res 2024; 19:550. [PMID: 39252111 PMCID: PMC11382417 DOI: 10.1186/s13018-024-05055-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by the degeneration, destruction, and excessive ossification of articular cartilage. The prevalence of OA is rising annually, concomitant with the aging global population and increasing rates of obesity. This condition imposes a substantial and escalating burden on individual health, healthcare systems, and broader social and economic frameworks. The etiology of OA is multifaceted and not fully understood. Current research suggests that the death of chondrocytes, encompassing mechanisms such as cellular apoptosis, pyroptosis, autophagy, ferroptosis and cuproptosis, contributes to both the initiation and progression of the disease. These cell death pathways not only diminish the population of chondrocytes but also exacerbate joint damage through the induction of inflammation and other deleterious processes. This paper delineates the morphological characteristics associated with various modes of cell death and summarizes current research results on the molecular mechanisms of different cell death patterns in OA. The objective is to review the advancements in understanding chondrocyte cell death in OA, thereby offering novel insights for potential clinical interventions.
Collapse
Affiliation(s)
- Mengqi Guan
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Qingyuan Yu
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Guohui Zhou
- Orthopedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Yan Wang
- Sino-Japanese Friendship Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Jianan Yu
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Wei Yang
- Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China
| | - Zhenhua Li
- Orthopedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, Jilin, China.
| |
Collapse
|
69
|
Tang Z, Feng H, Chen X, Shao S, Li C. SNORC knockdown alleviates inflammation, autophagy defect and matrix degradation of chondrocytes in osteoarthritis development. Mol Cell Biochem 2024; 479:2323-2335. [PMID: 37659033 DOI: 10.1007/s11010-023-04842-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
Excessive inflammation and autophagy defect of chondrocytes play important roles in the pathological process of osteoarthritis (OA). The present study aimed to clarify the roles of small novel rich in cartilage (SNORC) in these pathological changes of chondrocytes in OA. Bioinformatics analysis of GEO dataset GSE207881 displayed that SNORC was a potential biomarker for OA. As confirmed by quantitative real-time PCR, immunohistochemical staining and western blotting, SNORC was significantly up-regulated in cartilage of OA rat model and interleukin (IL)-1β-stimulated primary rat articular chondrocytes in contrast to their corresponding normal control. Knocking down SNORC in IL-1β-induced chondrocytes obviously suppressed the production of nitric oxide (NO), IL-6, tumor necrosis factor (TNF)-α and prostaglandin E2 (PGE2) to alleviate inflammation, and reduced the protein levels of a disintegrin and metalloproteinase with thrombospondin 5 (ADAMTS5) and matrix metallopeptidase (MMP)13 and elevated collagen type 2 alpha 1 (COL2A1) level to improve matrix degradation. Down-regulation of SNORC increased Beclin1 expression and LC3II/LC3I ratio, but suppressed p62 expression to restore impaired autophagy in IL-1β-induced chondrocytes. Moreover, down-regulating SNORC mitigated mitochondrial dysfunction and apoptosis in IL-1β-stimulated chondrocytes. Mechanically, SNORC simultaneously activated the phosphatidylinositol-3-kinase/serine threonine kinase (PI3K/AKT) and c-Jun N-terminal kinase (JNK)/c-Jun signaling pathway in the IL-1β-induced chondrocyte, while re-activating the PI3K and JNK signals abolished the suppressive effect of down-regulating SNORC on IL-1β-induced chondrocyte damage. In a word, SNORC knockdown alleviates inflammation, matrix degradation, autophagy defect and excessive apoptosis of chondrocytes during OA development via suppressing the PI3K and JNK signaling pathway.
Collapse
Affiliation(s)
- Zhifang Tang
- Clinical Medical College of Dali University, Dali, 671000, China
| | - Hanzhen Feng
- Clinical Medical College of Dali University, Dali, 671000, China
| | - Xusheng Chen
- Kunming Medical University, Kunming, 650500, China
| | - Shuiyan Shao
- Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Chuan Li
- Institute of Traumatology and Orthopedics, 920th Hospital of Joint Logistics Support Force, PLA, No.212 Daguan Road, Xishan District, Kunming, Yunnan, 650000, China.
| |
Collapse
|
70
|
Zhang Y, Zhao X, Shan L, Liu M, Zhang Z, Wang Z, Zhang X, Meng H, Song Y, Zhang W, Sang Z. Chronic Iodine Intake Excess Damages the Structure of Articular Cartilage and Epiphyseal Growth Plate. Biol Trace Elem Res 2024; 202:4078-4086. [PMID: 38060174 DOI: 10.1007/s12011-023-03985-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
This study aimed to explore the influence of excess iodine on the articular cartilage and epiphyseal growth plate in rats. Wistar rats (n = 200) were randomly divided into five groups with 40 rats in each: normal iodine (NI), 5-fold high iodine group (5HI), 10-fold high iodine group (10HI), 50-fold high iodine group (50HI), and 100-fold high iodine group (100HI). The rats were executed in 6 and 12 months. 24-h urinary iodine concentration (UIC) was monitored by arsenic-cerium catalytic spectrophotometry. The chemiluminescence method was used to determine the thyroid function. The pathological changes in the epiphyseal plate, articular cartilage, and thickness of the epiphyseal plate were observed. The mRNA expression of collagen II (ColII), collagen X, matrix metalloproteinase-13 (MMP-13), and fibroblast growth factor receptor 1 in articular chondrocytes was detected by RT-PCR. 24-h UIC increased as iodine intake increased. In the 12th month, there was a significant increase in serum sTSH and a decrease in serum FT4 in HI groups, compared to the NI group. There was a decrease in the number of proliferating cells in the epiphyseal plate and an increase in the number of mast cell layers. The chondrocytes appeared disorganized, and the tidal lines were disturbed or even broken. Growth plate thickness decreased with increasing iodine intake. Compared with the NI group, ColII and MMP-13 mRNA expression in chondrocytes in all HI groups significantly increased. Chronic iodine overdose increases the risk of hypothyroidism. Chronic iodine overdose leads to abnormal morphology of epiphyseal growth plates and articular cartilage, increasing the risk of osteoarthritis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Xin Zhao
- Department of Hand Microsurgery, Tianjin Hospital, Tianjin, China
| | - Le Shan
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Miao Liu
- Department of Comprehensive Office, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin, China
| | - Zixuan Zhang
- Department of Preventive Medicine, School of Public Health, Jilin University, Changchun City, China
| | - Zeji Wang
- Department of Medical Technology, Clinical Medical College of Tianjin Medical University, Tianjin, China
| | - Xinbao Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Haohao Meng
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Yan Song
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Wanqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China
| | - Zhongna Sang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Key Laboratory of Environmental Nutrition and Population Health, Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.
| |
Collapse
|
71
|
Zhang Z, Li B, Wu S, Yang Y, Wu B, Lai Q, Lai F, Mo F, Zhong Y, Wang S, Guo R, Zhang B. Bergenin protects against osteoarthritis by inhibiting STAT3, NF-κB and Jun pathways and suppressing osteoclastogenesis. Sci Rep 2024; 14:20292. [PMID: 39217193 PMCID: PMC11366014 DOI: 10.1038/s41598-024-71259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by articular cartilage destruction and subchondral bone reconstruction in the early stages. Bergenin (Ber) is a cytoprotective polyphenol found in many medicinal plants. It has been proven to have anti-inflammatory, antioxidant, and other biological activities, which may reveal its potential role in the treatment of OA. This study aimed to determine the potential efficacy of Ber in treating OA and explore the possible underlying mechanism through network pharmacology and validation experiments. The potential co-targets and processes of Ber and OA were predicted by using network pharmacology, including a Venn diagram for intersection targets, a protein‒protein interaction (PPI) network to obtain key potential targets, and GO and KEGG pathway enrichment to reveal the probable mechanism of action of Ber on OA. Subsequently, validation experiments were carried out to investigate the effects and mechanisms of Ber in treating OA in vitro and vivo. Ber suppressed IL-1β-induced chondrocyte apoptosis and extracellular matrix catabolism by inhibiting the STAT3, NF-κB and Jun signalling pathway in vitro. Furthermore, Ber suppressed the expression of osteoclast marker genes and RANKL-induced osteoclastogenesis. Ber alleviated the progression of OA in DMM-induced OA mice model. These results demonstrated the protective efficacy and potential mechanisms of Ber against OA, which suggested that Ber could be adopted as a potential therapeutic agent for treating OA.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Bo Li
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Shuqin Wu
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Yuxin Yang
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Binkang Wu
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Qi Lai
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Fuchong Lai
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Fengbo Mo
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
| | - Yufei Zhong
- Faculty of Jiangxi Medical College, Donghu District, Nanchang University, No.461 Bayi Road, Nanchang, 330006, Jiangxi, China
| | - Song Wang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| | - Runsheng Guo
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| | - Bin Zhang
- Department of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Department of Sports Medicine of Orthopedic Hospital, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, No. 17, Yongwaizheng Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
72
|
Lai C, Cheng X, Yuan T, Fang P, Qian H, Jiang H, Meng J, Zhao J, Bao N, Zhang L. A novel mechanism behind irreversible development of cartilage degradation driven articular cartilage defects revealed by rat model: The chain reaction initiated by extracellular vesicles delivered LOC102546541. Int Immunopharmacol 2024; 137:112467. [PMID: 38875997 DOI: 10.1016/j.intimp.2024.112467] [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: 04/04/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Articular cartilage defects (ACD) are injuries with a diameter greater than 3 mm, resulting from wear and tear on joints. When the diameter of the defect exceeds 6 mm, it can further damage the surrounding joint cartilage, causing osteoarthritis (OA). Try to explain why OA is an irreversible disease, we hypothesize that damaged articular chondrocytes (DAC) may have reduced capacities to repair cartilage because its extracellular vesicle (EVs) that might directly contribute to OA formation. METHODS In this study, DAC-EVs and AC-EVs were isolated using ultracentrifugation. Next-generation sequencing was employed to screen for a pathogenic long non-coding RNA (lncRNA). After verifying its function in vitro, the corresponding small interfering RNA (siRNA) was constructed and loaded into extracellular vesicles, which were then injected into the knee joint cavities of rats. RESULTS The results revealed that DAC-EVs packaged lncRNA LOC102546541 acts as a competitive endogenous RNA (ceRNA) of MMP13, down-regulating miR-632. Consequently, the function of MMP13 in degrading the extracellular matrix is enhanced, promoting the development of osteoarthritis. CONCLUSIONS This study uncovered a novel mode of OA pathogenesis using rat models, which DAC deliver pathogenic LOC102546541 packaged EVs to normal articular chondrocytes, amplifying the degradation of the extracellular matrix. Nonetheless, the functions of highly homologous human gene of LOC102546541 need to be verified in the future.
Collapse
Affiliation(s)
- Chengteng Lai
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xi Cheng
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Yuan
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Peng Fang
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hong Qian
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hui Jiang
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jia Meng
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jianning Zhao
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Nirong Bao
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Lei Zhang
- Department of Orthopaedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
73
|
Kan T, Tian Z, Sun L, Kong W, Yan R, Yu Z, Tian QW, Liu C. Quercetin-Loaded Zeolitic Imidazolate Framework-8 (ZIF-8) Nanoparticles Attenuate Osteoarthritis by Activating Autophagy via the Pi3k/Akt Signaling. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40444-40454. [PMID: 39058329 DOI: 10.1021/acsami.4c04649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Osteoarthritis treatment remains a significant clinical challenge. Quercetin, a natural flavonoid with anti-inflammatory and antiapoptotic properties, might be utilized to treat OA. However, poor water solubility and short joint retention duration limit its bioavailability and translation to clinical applications. A one-step self-assembly method was utilized to fabricate quercetin-loaded zeolitic imidazolate framework-8 (Qu@ZIF-8) nanoparticles using zinc ions, 2-methylimidazole, and quercetin. In vitro tests showed that Qu@ZIF-8 nanoparticles released pH-responsive agents into chondrocytes, effectively protecting them from interleukin (IL)-induced inflammation and apoptosis, thereby promoting cartilage anabolic activities. These underlying mechanisms revealed a remarkable increase of autophagy in IL-β-treated chondrocytes, followed by the inhibition of the Pi3k/Akt signaling pathway, which contributed to the protective effect of Qu @ZIF-8. By the establishment of medial meniscus instability (DMM) in OA mice, Qu@ZIF-8 substantially improved cartilage structural integrity and chondrocyte status, as well as attenuated OA progression. Importantly, Qu@ZIF-8 outperformed quercetin alone in the treatment of OA due to its control release. The combined research findings indicate that Qu@ZIF-8 shields chondrocytes from inflammation and apoptosis by activating autophagy and repressing the Pi3k/Akt pathway. This investigation may provide new insights for clinically extending the therapy of OA.
Collapse
Affiliation(s)
- Tianyou Kan
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Zhengtao Tian
- The State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200100, China
| | - Lin Sun
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Wei Kong
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Ruisi Yan
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Institution, Shanghai 200100 China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| | - Qi-Wei Tian
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Institution, Shanghai 200100 China
| | - Chenglei Liu
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Institution, Shanghai 200100, China
| |
Collapse
|
74
|
Ji Z, Ren X, Jin J, Ye X, Yu H, Fang W, Li H, Zhao Y, Tao S, Kong X, Cheng J, Shan Z, Chen J, Yao Q, Zhao F, Liu J. Injectable hydrogel encapsulating siMMP13 with anti-ROS and anti-apoptotic functions for osteoarthritis treatment. J Nanobiotechnology 2024; 22:466. [PMID: 39095867 PMCID: PMC11297633 DOI: 10.1186/s12951-024-02740-w] [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: 04/12/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive degeneration of articular cartilage, leading to pain, stiffness, and loss of joint function. The pathogenesis of OA involves multiple factors, including increased intracellular reactive oxygen species (ROS), enhanced chondrocyte apoptosis, and disturbances in cartilage matrix metabolism. These processes contribute to the breakdown of the extracellular matrix (ECM) and the loss of cartilage integrity, ultimately resulting in joint damage and dysfunction. RNA interference (RNAi) therapy has emerged as a promising approach for the treatment of various diseases, including hATTR and acute hepatic porphyria. By harnessing the natural cellular machinery for gene silencing, RNAi allows for the specific inhibition of target genes involved in disease pathogenesis. In the context of OA, targeting key molecules such as matrix metalloproteinase-13 (MMP13), which plays a critical role in cartilage degradation, holds great therapeutic potential. RESULTS In this study, we developed an innovative therapeutic approach for OA using a combination of liposome-encapsulated siMMP13 and NG-Monomethyl-L-arginine Acetate (L-NMMA) to form an injectable hydrogel. The hydrogel served as a delivery vehicle for the siMMP13, allowing for sustained release and targeted delivery to the affected joint. Experiments conducted on destabilization of the medial meniscus (DMM) model mice demonstrated the therapeutic efficacy of this composite hydrogel. Treatment with the hydrogel significantly inhibited the degradation of cartilage matrix, as evidenced by histological analysis showing preserved cartilage structure and reduced loss of proteoglycans. Moreover, the hydrogel effectively suppressed intracellular ROS accumulation in chondrocytes, indicating its anti-oxidative properties. Furthermore, it attenuated chondrocyte apoptosis, as demonstrated by decreased levels of apoptotic markers. CONCLUSION In summary, the injectable hydrogel containing siMMP13, endowed with anti-ROS and anti-apoptotic properties, may represent an effective therapeutic strategy for osteoarthritis in the future.
Collapse
Affiliation(s)
- Zhongyin Ji
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Department of Orthopedics Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P.R. China
| | - Xiaobin Ren
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, P.R. China
| | - Jiayan Jin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Xin Ye
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Hao Yu
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, P.R. China
| | - Wenhan Fang
- College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, P.R. China
| | - Hui Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Yihao Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Xiangxi Kong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Jiao Cheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Zhi Shan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China
| | - Qingqing Yao
- School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Xi Road, Wenzhou, 325027, Zhejiang, P.R. China.
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, No. 3, Qingchun Road East, Hangzhou, 310016, P.R. China.
| |
Collapse
|
75
|
Kuang S, Liu Z, Liu L, Fu X, Sheng W, Hu Z, Lin C, He Q, Chen J, Gao S. Polygonatum sibiricum polysaccharides protect against knee osteoarthritis by inhibiting the TLR2/NF-κB signaling pathway in vivo and in vitro. Int J Biol Macromol 2024; 274:133137. [PMID: 38901508 DOI: 10.1016/j.ijbiomac.2024.133137] [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: 03/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
Polygonatum sibiricum polysaccharides (PSP), the primary constituent of Polygonatum sibiricum, have been shown to exhibit a wide range of pharmacological effects, but their impact on osteoarthritis (OA) remains unclear. The objective of this study was to investigate the protective effects of PSP against OA and to elucidate its underlying molecular mechanism. In our in vitro experiments, PSP not only inhibited the IL-1β-induced inflammatory responses and the nuclear factor kappa-B (NF-κB) signaling pathway in chondrocytes but also regulated the cartilage matrix metabolism. In addition, we detected 394 significantly differentially expressed genes through RNA-seq analysis on PSP-intervened chondrocytes, and the toll-like receptor 2 (TLR2) was identified as the most important feature by functional network analysis and qRT-PCR. It was also revealed that PSP treatment significantly reversed the IL-1-induced up-regulation of TLR2 expression in chondrocytes, while TLR2 overexpression partially inhibited the regulatory effects of PSP on inflammation, NF-κB signaling pathway and matrix metabolism. In our in vivo experiments, PSP treatment alleviated the development of destabilization of medial meniscus (DMM)-induced OA in mouse knee joints, inhibited the DMM-induced activation of the TLR2/NF-κB signaling pathway in mouse knee joint cartilage, and reduced the serum levels of inflammatory cytokines. In conclusion, PSP exerts its anti-inflammatory, matrix synthesis-promoting and matrix catabolism-suppressing effects in knee OA by inhibiting the TLR2/NF-κB signaling pathway, suggesting that PSP may be potentially targeted as a novel all-natural, low-toxicity drug for OA prevention and treatment.
Collapse
Affiliation(s)
- Shida Kuang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhewen Liu
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Lumei Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xinying Fu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Wen Sheng
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Zongren Hu
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Chengxiong Lin
- Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Qinghu He
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Medicine, Huaihua, Hunan 418000, China.
| | - Jisong Chen
- Hunan University of Medicine, Huaihua, Hunan 418000, China.
| | - Shuguang Gao
- Department of Orthopaedics, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
76
|
Oo WM. Prospects of Disease-Modifying Osteoarthritis Drugs. Rheum Dis Clin North Am 2024; 50:483-518. [PMID: 38942581 DOI: 10.1016/j.rdc.2024.03.003] [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] [Indexed: 06/30/2024]
Abstract
Osteoarthritis (OA) causes a massive disease burden with a global prevalence of nearly 23% in 2020 and an unmet need for adequate treatment, given a lack of disease-modifying drugs (DMOADs). The author reviews the prospects of active DMOAD candidates in the phase 2/3 clinical trials of drug development pipeline based on key OA pathogenetic mechanisms directed to inflammation-driven, bone-driven, and cartilage-driven endotypes. The challenges and possible research opportunities are stated in terms of the formulation of a research question known as the PICO approach: (1) population, (2) interventions, (3) comparison or placebo, and (4) outcomes.
Collapse
Affiliation(s)
- Win Min Oo
- Department of Physical Medicine and Rehabilitation, Mandalay General Hospital, University of Medicine, Mandalay, Mandalay, Myanmar; Rheumatology Department, Royal North Shore Hospital, Institute of Bone and Joint Research, Kolling Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
77
|
Chang YH, Wu KC, Hsu CJ, Tu TC, Liu MC, Chiang RYS, Ding DC. Therapeutic Potential of Olfactory Ensheathing Cells and Adipose-Derived Stem Cells in Osteoarthritis: Insights from Preclinical Studies. Cells 2024; 13:1250. [PMID: 39120281 PMCID: PMC11311847 DOI: 10.3390/cells13151250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Olfactory-ensheathing cells (OECs) are known for their role in neuronal regeneration and potential to promote tissue repair. Adipose-derived stem cells (ADSCs), characterized by mesenchymal stem cell (MSC) traits, display a fibroblast-like morphology and express MSC surface markers, making them suitable for regenerative therapies for osteoarthritis (OA). In this study, OECs and ADSCs were derived from tissues and characterized for their morphology, surface marker expression, and differentiation capabilities. Collagenase-induced OA was created in 10-week-old C57BL/6 mice, followed by intra-articular injections of ADSCs (1 × 105), OECs (1 × 105), or a higher dose of OECs (5 × 105). Therapeutic efficacy was evaluated using rotarod performance tests, MRI, histology, and immunohistochemistry. Both cell types exhibited typical MSC characteristics and successfully differentiated into adipocytes, osteoblasts, and chondrocytes, confirmed by gene expression and staining. Transplantation significantly improved rotarod performance and preserved cartilage integrity, as seen in MRI and histology, with reduced cartilage destruction and increased chondrocytes. Immunohistochemistry showed elevated type II collagen and aggrecan in treated joints, indicating hyaline cartilage formation, and reduced MMP13 and IL-1β expression, suggesting decreased inflammation and catabolic activity. These findings highlight the regenerative potential of OECs and ADSCs in treating OA by preserving cartilage, promoting chondrocyte proliferation, and reducing inflammation. Further research is needed to optimize delivery methods and evaluate long-term clinical outcomes.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Chih-Jung Hsu
- Top Medical Biomedical Co., Ltd., Yilan City 260, Taiwan; (C.-J.H.); (T.-C.T.); (M.-C.L.)
| | - Tsui-Chin Tu
- Top Medical Biomedical Co., Ltd., Yilan City 260, Taiwan; (C.-J.H.); (T.-C.T.); (M.-C.L.)
| | - Mei-Chun Liu
- Top Medical Biomedical Co., Ltd., Yilan City 260, Taiwan; (C.-J.H.); (T.-C.T.); (M.-C.L.)
| | - Raymond Yuh-Shyan Chiang
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
78
|
Hu YC, Huang TC, Huang LW, Cheng HL, Hsieh BS, Chang KL. S-Equol Ameliorates Menopausal Osteoarthritis in Rats through Reducing Oxidative Stress and Cartilage Degradation. Nutrients 2024; 16:2364. [PMID: 39064807 PMCID: PMC11280421 DOI: 10.3390/nu16142364] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease leading to articular cartilage destruction. Menopausal and postmenopausal women are susceptible to both OA and osteoporosis. S-equol, a soy isoflavone-derived molecule, is known to reduce osteoporosis in estrogen-deficient mice, but its role in OA remains unknown. This study aimed to explore the effect of S-equol on different degrees of menopausal OA in female Sprague-Dawley (SD) rats induced by estrogen deficiency caused by bilateral ovariectomy (OVX) combined with intra-articular injection of mono-iodoacetate (MIA). Knee joint histopathological change; serum biomarkers of bone turnover, including N-terminal propeptide of type I procollagen (PINP), C-terminal telopeptide of type I collagen (CTX-I) and N-terminal telopeptide of type I collagen (NTX-I); the cartilage degradation biomarkers hyaluronic acid (HA) and N-terminal propeptide of type II procollagen (PIINP); and the matrix-degrading enzymes matrix metalloproteinases (MMP)-1, MMP-3 and MMP-13, as well as the oxidative stress-inducing molecules nitric oxide (NO) and hydrogen peroxide (H2O2), were assessed for evaluation of OA progression after S-equol supplementation for 8 weeks. The results showed that OVX without or with MIA injection induced various severity levels of menopausal OA by increasing pathological damage, oxidative stress, and cartilage matrix degradation to various degrees. Moreover, S-equol supplementation could significantly reduce these increased biomarkers in different severity levels of OA. This indicates that S-equol can lessen menopausal OA progression by reducing oxidative stress and the matrix-degrading enzymes involved in cartilage degradation.
Collapse
Affiliation(s)
- Yu-Chen Hu
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Tzu-Ching Huang
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Li-Wen Huang
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Hsiao-Ling Cheng
- Department of Pharmacy, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802511, Taiwan;
| | - Bau-Shan Hsieh
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Kee-Lung Chang
- Department of Biochemistry, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (Y.-C.H.); (T.-C.H.); (B.-S.H.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
79
|
de la Fuente M, Delgado D, Beitia M, Barreda-Gómez G, Acera A, Sanchez M, Vecino E. Validation of a rapid collagenase activity detection technique based on fluorescent quenched gelatin with synovial fluid samples. BMC Biotechnol 2024; 24:50. [PMID: 39030513 PMCID: PMC11264812 DOI: 10.1186/s12896-024-00869-y] [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: 02/24/2024] [Accepted: 06/13/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Measuring collagenase activity is crucial in the field of joint health and disease management. Collagenases, enzymes responsible for collagen degradation, play a vital role in maintaining the balance between collagen synthesis and breakdown in joints. Dysregulation of collagenase activity leads to joint tissue degradation and diseases such as rheumatoid arthritis and osteoarthritis. The development of methods to measure collagenase activity is essential for diagnosis, disease severity assessment, treatment monitoring, and identification of therapeutic targets. RESULTS This study aimed to validate a rapid collagenase activity detection technique using synovial fluid samples. Antibody microarray analysis was initially performed to quantify the levels of matrix metalloproteinase-9 (MMP-9), a major collagenase in joints. Subsequently, the developed gelatin-based test utilizing fluorescence measurement was used to determine collagenase activity. There was a significant correlation between the presence of MMP-9 and collagenase activity. In addition, Lower Limit of Detection and Upper Limit of Detection can be preliminary estimated as 8 ng/mL and 48 ng/mL respectively. CONCLUSIONS The developed technique offers a potential point-of-care assessment of collagenase activity, providing real-time information for clinicians and researchers. By accurately quantifying collagenase activity, healthcare professionals can optimize patient care, improve treatment outcomes, and contribute to the understanding and management of joint-related disorders. Further research and validation are necessary to establish the full potential of this rapid collagenase activity detection method in clinical practice.
Collapse
Affiliation(s)
- Miguel de la Fuente
- Department of Cell Biology and Histology, Experimental Ophthalmo-Biology Group (GOBE, www.ehu.eus/gobe), University of the Basque Country UPV/EHU, Leioa, 48940, Spain
| | - Diego Delgado
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, 01008, Spain
| | - Maider Beitia
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, 01008, Spain
| | | | - Arantxa Acera
- Department of Cell Biology and Histology, Experimental Ophthalmo-Biology Group (GOBE, www.ehu.eus/gobe), University of the Basque Country UPV/EHU, Leioa, 48940, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48001, Spain
| | - Mikel Sanchez
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, 01008, Spain
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, Vitoria-Gasteiz, 01008, Spain
| | - Elena Vecino
- Department of Cell Biology and Histology, Experimental Ophthalmo-Biology Group (GOBE, www.ehu.eus/gobe), University of the Basque Country UPV/EHU, Leioa, 48940, Spain.
| |
Collapse
|
80
|
Huang J, Ren Q, Jiao L, Niu S, Liu C, Zhou J, Wu L, Yang Y. TMF suppresses chondrocyte hypertrophy in osteoarthritic cartilage by mediating the FOXO3a/BMPER pathway. Exp Ther Med 2024; 28:283. [PMID: 38800044 PMCID: PMC11117099 DOI: 10.3892/etm.2024.12571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Osteoarthritis (OA) is a disease of the joints, characterized by chronic inflammation, cartilage destruction and extracellular matrix (ECM) remodeling. Aberrant chondrocyte hypertrophy promotes cartilage destruction and OA development. Collagen X, the biomarker of chondrocyte hypertrophy, is upregulated by runt-related transcription factor 2 (Runx2), which is mediated by the bone morphogenetic protein 4 (BMP4)/Smad1 signaling pathway. BMP binding endothelial regulator (BMPER), a secreted glycoprotein, acts as an agonist of BMP4. 5,7,3',4'-tetramethoxyflavone (TMF) is a natural flavonoid derived from Murraya exotica L. Results of our previous study demonstrated that TMF exhibits chondroprotective effects against OA development through the activation of Forkhead box protein O3a (FOXO3a) expression. However, whether TMF suppresses chondrocyte hypertrophy through activation of FOXO3a expression and inhibition of BMPER/BMP4/Smad1 signaling remains unknown. Results of the present study revealed that TMF inhibited collagen X and Runx2 expression, inhibited BMPER/BMP4/Smad1 signaling, and activated FOXO3a expression; thus, protecting against chondrocyte hypertrophy and OA development. However, BMPER overexpression and FOXO3a knockdown impacted the protective effects of TMF. Thus, TMF inhibited chondrocyte hypertrophy in OA cartilage through mediating the FOXO3a/BMPER signaling pathway.
Collapse
Affiliation(s)
- Jishang Huang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Qun Ren
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Linhui Jiao
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Shuo Niu
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Chenghong Liu
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Juan Zhou
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Yadong Yang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
81
|
Zhou D, Wei Y, Sheng S, Wang M, Lv J, Zhao B, Chen X, Xu K, Bai L, Wu Y, Song P, Cao L, Zhou F, Zhang H, Shi Z, Su J. MMP13-targeted siRNA-loaded micelles for diagnosis and treatment of posttraumatic osteoarthritis. Bioact Mater 2024; 37:378-392. [PMID: 38689658 PMCID: PMC11059470 DOI: 10.1016/j.bioactmat.2024.04.010] [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: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Posttraumatic osteoarthritis (PTOA) patients are often diagnosed by X-ray imaging at a middle-late stage when drug interventions are less effective. Early PTOA is characterized by overexpressed matrix metalloprotease 13 (MMP13). Herein, we constructed an integrated diagnosis and treatment micelle modified with MMP13 enzyme-detachable, cyanine 5 (Cy5)-containing PEG, black hole quencher-3 (BHQ3), and cRGD ligands and loaded with siRNA silencing MMP13 (siM13), namely ERMs@siM13. ERMs@siM13 could be cleaved by MMP13 in the diseased cartilage tissues to detach the PEG shell, causing cRGD exposure. Accordingly, the ligand exposure promoted micelle uptake by the diseased chondrocytes by binding to cell surface αvβ3 integrin, increasing intracellular siM13 delivery for on-demand MMP13 downregulation. Meanwhile, the Cy5 fluorescence was restored by detaching from the BHQ3-containing micelle, precisely reflecting the diseased cartilage state. In particular, the intensity of Cy5 fluorescence generated by ERMs@siM13 that hinged on the MMP13 levels could reflect the PTOA severity, enabling the physicians to adjust the therapeutic regimen. Finally, in the murine PTOA model, ERMs@siM13 could diagnose the early-stage PTOA, perform timely interventions, and monitor the OA progression level during treatment through a real-time detection of MMP13. Therefore, ERMs@siM13 represents an appealing approach for early-stage PTOA theranostics.
Collapse
Affiliation(s)
- Dongyang Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Shihao Sheng
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Miaomiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- Department of Rehabilitation Medicine, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
| | - Bowen Zhao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiao Chen
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Peiran Song
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Hao Zhang
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhongmin Shi
- Department of Orthopedics, Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai), Shanghai University, Shanghai, 200444, China
- Department of Orthopedic, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
82
|
James JJ, Sandhya KV, Sridhar KN, Sudarson S, Basavaraj BV, Bharath S. Proteomic Characterization of Human Placenta: Insights into Potential Therapeutic Applications for Osteoarthritis. AAPS PharmSciTech 2024; 25:139. [PMID: 38890179 DOI: 10.1208/s12249-024-02851-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024] Open
Abstract
Biologics have become increasingly prominent as therapeutics in recent years due to their innate immune-privileged nature, biocompatibility, and high levels of protein biofactors. The aim of the study is to characterise the biologic, lyophilized human placenta (LHP) and explore its therapeutic potential for osteoarthritis (OA). The presence of six bioactive constituents that regulate cell-extracellular matrix interaction was identified by liquid chromatography coupled to electrospray ionization and quadrupole time-of-flight mass spectrometry (LC-ESI-QTOF/MS). Metalloproteinase inhibitor 3 (TIMP3), alpha-1 anti-trypsin (a1AT), basic fibroblast growth factor (bFGF), and transforming growth factor β1 (TGFβ1) were detected and quantified using ELISA. The total protein content present in LHP by Bradford assay was found to be 409.35 ± 0.005 μg/ml. The analytical techniques such as Attenuated Total Reflectance-Fourier Transform Infrared spectroscopy (ATR-FTIR), solid state carbon-13 Nuclear Magnetic Resonance (ssC13 NMR) spectroscopy, and Differential Scanning Calorimetry (DSC) revealed the secondary structure and conformational stability of LHP. X-Ray diffraction (XRD) studies showed its amorphous nature. Bioactivity assessment of LHP was performed in human keratinocytes (HaCaT) and human dermal fibroblasts (HDF) by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The LHP was highly proliferative against skin cells and non-toxic, based on the findings of the bioactivity assay. LHP has the potential to be used as a therapeutic agent for OA, as its characterisation unveiled its physical stability, significant concentration of bioactive components that are pertinent to cartilage repair and its conformational stability.
Collapse
Affiliation(s)
- Jithu Jerin James
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, University House, Gnanagangothri Campus, New BEL Road, MSR Nagar, Bengaluru, 560054, India
| | - K V Sandhya
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, University House, Gnanagangothri Campus, New BEL Road, MSR Nagar, Bengaluru, 560054, India.
| | - K N Sridhar
- Cancyte Technologies Pvt Ltd, Sri Shankara Research Centre, Rangadore Memorial Hospital, Shankarapuram, Bengaluru, India
| | - S Sudarson
- Cancyte Technologies Pvt Ltd, Sri Shankara Research Centre, Rangadore Memorial Hospital, Shankarapuram, Bengaluru, India
| | - B V Basavaraj
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, University House, Gnanagangothri Campus, New BEL Road, MSR Nagar, Bengaluru, 560054, India
| | - S Bharath
- Department of Pharmaceutics, Faculty of Pharmacy, MS Ramaiah University of Applied Sciences, University House, Gnanagangothri Campus, New BEL Road, MSR Nagar, Bengaluru, 560054, India
| |
Collapse
|
83
|
Hamm P, Meinel L, Driessen MD. An Introductory Guide to Protease Sensitive Linker Design Using Matrix Metalloproteinase 13 as an Example. ACS Biomater Sci Eng 2024; 10:3693-3706. [PMID: 38813796 DOI: 10.1021/acsbiomaterials.4c00407] [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] [Indexed: 05/31/2024]
Abstract
Proteases play a crucial role, not only in physiological, but also in pathological processes, such as cancer, inflammation, arthritis, Alzheimer's, and infections, to name but a few. Their ability to cleave peptides can be harnessed for a broad range of biotechnological purposes. To do this efficiently, it is essential to find an amino acid sequence that meets the necessary requirements, including interdependent factors like specificity, selectivity, cleavage kinetics, or synthetic accessibility. Cleavage sequences from natural substrates of the protease may not be optimal in terms of specificity and selectivity, which is why these frequently require arduous and sometimes unsuccessful optimization such as by iterative exchange of single amino acids. Hence, here we describe the systematic design of protease sensitive linkers (PSLs)─peptide sequences specifically cleaved by a target protease─guided by the mass spectrometry based determination of target protease specific cleavage sites from a proteome-based peptide library. It includes a procedure for identifying bespoke PSL sequences, their optimization, synthesis, and validation and introduces a program that can indicate potential cleavage sites by hundreds of enzymes in any arbitrary amino acid sequence. Thereby, we provide an introduction to PSL design, illustrated by the example of matrix metalloproteinase 13 (MMP13). This introduction can serve as a guide and help to greatly accelerate the development and use of protease-sensitive linkers in diverse applications.
Collapse
Affiliation(s)
- Prisca Hamm
- Institute for Pharmacy and Food Chemistry, University of Würzburg, 97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Würzburg, 97074 Würzburg, Germany
- Helmholtz-Institute for RNA-Based Infection Research (HIRI), 97070 Würzburg, Germany
| | - Marc D Driessen
- Institute of Molecular Medicine I, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
- Department for Oral and Craniomaxillofacial and Plastic Surgery, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
84
|
Szala D, Kopańska M, Trojniak J, Jabłoński J, Hanf-Osetek D, Snela S, Zawlik I. The Role of MicroRNAs in the Pathophysiology of Osteoarthritis. Int J Mol Sci 2024; 25:6352. [PMID: 38928059 PMCID: PMC11204066 DOI: 10.3390/ijms25126352] [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: 05/05/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Worldwide, osteoarthritis (OA) is the most common cause of joint pain in older people. Many factors contribute to osteoarthritis' development and progression, including secondary osteoarthritis' underlying causes. It is important to note that osteoarthritis affects all four tissues: cartilage, bone, joint capsule, and articular apparatus. An increasingly prominent area of research in osteoarthritis regulation is microRNAs (miRNAs), a small, single-stranded RNA molecule that controls gene expression in eukaryotes. We aimed to assess and summarize current knowledge about the mechanisms of the action of miRNAs and their clinical significance. Osteoarthritis (OA) is affected by the interaction between miRNAs and inflammatory processes, as well as cartilage metabolism. MiRNAs also influence cartilage cell apoptosis, contributing to the degradation of the cartilage in OA. Studies have shown that miRNAs may have both an inhibitory and promoting effect on osteoporosis progression through their influence on molecular mechanisms. By identifying these regulators, targeted treatments for osteoarthritis may be developed. In addition, microRNA may also serve as a biomarker for osteoarthritis. By using these biomarkers, the disease could be detected faster, and early intervention can be instituted to prevent mobility loss and slow deterioration.
Collapse
Affiliation(s)
| | - Marta Kopańska
- Department of Pathophysiology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Julia Trojniak
- Student Research Club “Reh-Tech”, Medical College of Rzeszow University, 35-959 Rzeszow, Poland;
| | - Jarosław Jabłoński
- Faculty of Orthopaedic and Reumatology, Institute of Medical Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland; (J.J.); (D.H.-O.); (S.S.)
- Orthopaedics and Traumatology Clinic, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Dorota Hanf-Osetek
- Faculty of Orthopaedic and Reumatology, Institute of Medical Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland; (J.J.); (D.H.-O.); (S.S.)
- Orthopaedics and Traumatology Clinic, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Sławomir Snela
- Faculty of Orthopaedic and Reumatology, Institute of Medical Sciences, Collegium Medicum, University of Rzeszow, 35-959 Rzeszow, Poland; (J.J.); (D.H.-O.); (S.S.)
- Orthopaedics and Traumatology Clinic, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Izabela Zawlik
- Department of General Genetics, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland;
| |
Collapse
|
85
|
Morris JL, Letson HL, McEwen PC, Dobson GP. Adenosine, lidocaine, and magnesium therapy augments joint tissue healing following experimental anterior cruciate ligament rupture and reconstruction. Bone Joint Res 2024; 13:279-293. [PMID: 38843878 PMCID: PMC11156504 DOI: 10.1302/2046-3758.136.bjr-2023-0360.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/10/2024] Open
Abstract
Aims Adenosine, lidocaine, and Mg2+ (ALM) therapy exerts differential immuno-inflammatory responses in males and females early after anterior cruciate ligament (ACL) reconstruction (ACLR). Our aim was to investigate sex-specific effects of ALM therapy on joint tissue repair and recovery 28 days after surgery. Methods Male (n = 21) and female (n = 21) adult Sprague-Dawley rats were randomly divided into ALM or Saline control treatment groups. Three days after ACL rupture, animals underwent ACLR. An ALM or saline intravenous infusion was commenced prior to skin incision, and continued for one hour. An intra-articular bolus of ALM or saline was also administered prior to skin closure. Animals were monitored to 28 days, and joint function, pain, inflammatory markers, histopathology, and tissue repair markers were assessed. Results Despite comparable knee function, ALM-treated males had reduced systemic inflammation, synovial fluid angiogenic and pro-inflammatory mediators, synovitis, and fat pad fibrotic changes, compared to controls. Within the ACL graft, ALM-treated males had increased expression of tissue repair markers, decreased inflammation, increased collagen organization, and improved graft-bone healing. In contrast to males, females had no evidence of persistent systemic inflammation. Compared to controls, ALM-treated females had improved knee extension, gait biomechanics, and elevated synovial macrophage inflammatory protein-1 alpha (MIP-1α). Within the ACL graft, ALM-treated females had decreased inflammation, increased collagen organization, and improved graft-bone healing. In articular cartilage of ALM-treated animals, matrix metalloproteinase (MMP)-13 expression was blunted in males, while in females repair markers were increased. Conclusion At 28 days, ALM therapy reduces inflammation, augments tissue repair patterns, and improves joint function in a sex-specific manner. The study supports transition to human safety trials.
Collapse
Affiliation(s)
- Jodie L. Morris
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Hayley L. Letson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Peter C. McEwen
- Orthopaedic Research Institute of Queensland, Townsville, Australia
| | - Geoffrey P. Dobson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| |
Collapse
|
86
|
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
|
87
|
Yan W, Li Y, Xie S, Tao WA, Hu J, Liu H, Zhang G, Liu F, Nie Y, Chen X, Zhang X, Liu Y, Wei D, Ma C, Zhang H, Xu H, Wang S. Chondrocyte-Targeted Delivery System of Sortase A-Engineered Extracellular Vesicles Silencing MMP13 for Osteoarthritis Therapy. Adv Healthc Mater 2024; 13:e2303510. [PMID: 38545904 DOI: 10.1002/adhm.202303510] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/21/2024] [Indexed: 04/09/2024]
Abstract
Targeted drug delivery and the reduction of off-target effects are crucial for the promising clinical application of nucleic acid drugs. To address this challenge, a new approach for treating osteoarthritis (OA) that accurately delivers antisense oligonucleotides (ASO) targeting matrix metalloproteinase-13 (ASO-MMP13) to chondrocytes, is developed. Small extracellular vesicles (exos) are ligated with chondrocyte affinity peptide (CAP) using Sortase A and subsequently incubated with cholesterol-modified ASO-MMP13 to construct a chondrocyte-targeted drug delivery exo (CAP-exoASO). Compared with exos without CAP (ExoASO), CAP-exoASOs attenuate IL-1β-induced chondrocyte damage and prolong the retention time of ASO-MMP13 in the joint without distribution in major organs following intra-articular injection. Notably, CAP-exoASOs decrease MMP13 expression (P < 0.001) and upregulate COL2A1 expression (P = 0.006), resulting in reorganization of the cartilage matrix and alleviation of progression in the OA model. Furthermore, the Osteoarthritis Research Society International (OARSI) score of articular cartilage tissues treated with CAP-exoASO is comparable with that of healthy rats (P = 0.148). A mechanistic study demonstrates that CAP-exoASO may reduce inflammation by suppressing the IL-17 and TNF signaling pathways. Based on the targeted delivery effect, CAP-exoASOs successfully accomplish cartilage repair and have considerable potential for development as a promising therapeutic modality for satisfactory OA therapy.
Collapse
Affiliation(s)
- Wenjing Yan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Ying Li
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Department of Epidemiology, School of Public Health of Suzhou University, Suzhou, Jiangsu, 215127, China
| | - Shuqian Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - W Andy Tao
- Departments of Chemistry and Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Jing Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Haohan Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Guiyuan Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Fengying Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Yamei Nie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Xue Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Xing Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| | - Yufeng Liu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Dong Wei
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Changyan Ma
- Department of Medical Genetics, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hao Zhang
- EVLiXiR Biotech Inc., Nanjing, Jiangsu, 210032, China
| | - Hongtao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210003, China
| |
Collapse
|
88
|
Petta D, D'Arrigo D, Salehi S, Talò G, Bonetti L, Vanoni M, Deabate L, De Nardo L, Dubini G, Candrian C, Moretti M, Lopa S, Arrigoni C. A personalized osteoarthritic joint-on-a-chip as a screening platform for biological treatments. Mater Today Bio 2024; 26:101072. [PMID: 38757057 PMCID: PMC11097088 DOI: 10.1016/j.mtbio.2024.101072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Osteoarthritis (OA) is a highly disabling pathology, characterized by synovial inflammation and cartilage degeneration. Orthobiologics have shown promising results in OA treatment thanks to their ability to influence articular cells and modulate the inflammatory OA environment. Considering their complex mechanism of action, the development of reliable and relevant joint models appears as crucial to select the best orthobiologics for each patient. The aim of this study was to establish a microfluidic OA model to test therapies in a personalized human setting. The joint-on-a-chip model included cartilage and synovial compartments, containing hydrogel-embedded chondrocytes and synovial fibroblasts, separated by a channel for synovial fluid. For the cartilage compartment, a Hyaluronic Acid-based matrix was selected to preserve chondrocyte phenotype. Adding OA synovial fluid induced the production of inflammatory cytokines and degradative enzymes, generating an OA microenvironment. Personalized models were generated using patient-matched cells and synovial fluid to test the efficacy of mesenchymal stem cells on OA signatures. The patient-specific models allowed monitoring changes induced by cell injection, highlighting different individual responses to the treatment. Altogether, these results support the use of this joint-on-a-chip model as a prognostic tool to screen the patient-specific efficacy of orthobiologics.
Collapse
Affiliation(s)
- Dalila Petta
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
| | - Daniele D'Arrigo
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- ISBE-SYSBIO Centre of Systems Biology, Milan, Italy at Department of Biotechnology and Biosciences, Università Degli Studi di Milano Bicocca, Piazza Della Scienza 2, 20126, Milan, Italy
| | - Shima Salehi
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Marco Vanoni
- ISBE-SYSBIO Centre of Systems Biology, Milan, Italy at Department of Biotechnology and Biosciences, Università Degli Studi di Milano Bicocca, Piazza Della Scienza 2, 20126, Milan, Italy
| | - Luca Deabate
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Gabriele Dubini
- Department of Chemistry, Materials and Chemical Engineering G.Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Christian Candrian
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Via Belgioioso 173, 20157, Milan, Italy
| | - Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Via Chiesa, 5, 6500, Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale, Via Tesserete 46, 6900, Lugano, Switzerland
- Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana (USI), Via Buffi 13, 6900, Lugano, Switzerland
| |
Collapse
|
89
|
Wang X, Zhou Y, Luo C, Zhao J, Ji Y, Wang Z, Zheng P, Li D, Shi Y, Nishiura A, Matsumoto N, Honda Y, Xu B, Huang F. Senolytics ameliorate the failure of bone regeneration through the cell senescence-related inflammatory signalling pathway. Biomed Pharmacother 2024; 175:116606. [PMID: 38670048 DOI: 10.1016/j.biopha.2024.116606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Stress-induced premature senescent (SIPS) cells induced by various stresses deteriorate cell functions. Dasatinib and quercetin senolytics (DQ) can alleviate several diseases by eliminating senescent cells. α-tricalcium phosphate (α-TCP) is a widely used therapeutic approach for bone restoration but induces bone formation for a comparatively long time. Furthermore, bone infection exacerbates the detrimental prognosis of bone formation during material implant surgery due to oral cavity bacteria and unintentional contamination. It is essential to mitigate the inhibitory effects on bone formation during surgical procedures. Little is known that DQ improves bone formation in Lipopolysaccharide (LPS)-contaminated implants and its intrinsic mechanisms in the study of maxillofacial bone defects. This study aims to investigate whether the administration of DQ ameliorates the impairments on bone repair inflammation and contamination by eliminating SIPS cells. α-TCP and LPS-contaminated α-TCP were implanted into Sprague-Dawley rat calvaria bone defects. Simultaneously, bone formation in the bone defects was investigated with or without the oral administration of DQ. Micro-computed tomography and hematoxylin-eosin staining showed that senolytics significantly enhanced bone formation at the defect site. Histology and immunofluorescence staining revealed that the levels of p21- and p16-positive senescent cells, inflammation, macrophages, reactive oxygen species, and tartrate-resistant acid phosphatase-positive cells declined after administering DQ. DQ could partially alleviate the production of senescent markers and senescence-associated secretory phenotypes in vitro. This study indicates that LPS-contaminated α-TCP-based biomaterials can induce cellular senescence and hamper bone regeneration. Senolytics have significant therapeutic potential in reducing the adverse osteogenic effects of biomaterial-related infections and improving bone formation capacity.
Collapse
Affiliation(s)
- Xinchen Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Yue Zhou
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan; Department of Stomatological Research Center, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Chuyi Luo
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Jianxin Zhao
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Yuna Ji
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zheng Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pengchao Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dingji Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhan Shi
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Aki Nishiura
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Naoyuki Matsumoto
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Yoshitomo Honda
- Department of Oral Anatomy, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan.
| | - Baoshan Xu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Fang Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
90
|
Cheng L, Wang K, Chang S, Tan Y, He B. Effects of platelet-rich plasma combined with isometric quadriceps contraction on cartilage in a rat model of knee osteoarthritis. Regen Ther 2024; 26:469-477. [PMID: 39070125 PMCID: PMC11283084 DOI: 10.1016/j.reth.2024.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Intra-articular injection of platelet-rich plasma (PRP) or isometric contraction of quadriceps (ICQ) has shown positive effects in patients with knee osteoarthritis (KOA). However, the synergistic effect of combining PRP and ICQ intervention (joint intervention) on cartilage repair has not been validated. Thus, this study aimed to explore the reparative effects of joint intervention on cartilage in a KOA rat model. METHODS Fifty-four 2-month-old female Sprague-Dawley rats were randomly divided into the control group (CG, n = 6) and model group (injected with sodium iodoacetate, n = 48). After 1 week, six rats from the model group were randomly selected for validation. The remaining 42 rats were further divided into seven groups: PRP group (PRPG), ICQ group (ICQG), joint intervention group (JIG), normal saline group (NSG), acupuncture group (AG), normal saline and acupuncture group (NSAG) and model blank group (MBG). The intervention lasted for 4 weeks, with PRPG and JIG receiving PRP injections (twice) and ICQG and JIG undergoing ICQ (five times per week, 15 min each session). RESULTS Histological staining with haematoxylin and eosin as well as transmission electron microscopy revealed severe cartilage damage in MBG, AG, NSAG and NSG, followed by PRPG and ICQG. JIG exhibited a more intact cartilage structure. Compared with JIG, the Mankin scores increased remarkably in PRPG, ICQG, AG, NSAG and NSG (P < 0.01). Relative mRNA expression levels showed the upregulation of IL-1β in ICQG, NSAG and NSG compared with JIG (P < 0.05) and the upregulation of IL-6, IL-18 and MMP-13 in AG and NSAG (P < 0.05). Compared with PRPG, IL-1β and IL-6 were upregulated in ICQG, AG, NSAG and NSG (P < 0.05). In addition, IL-18 was upregulated in AG (P < 0.01), and IL-18, MMP-13 and TNF-α were upregulated in NSAG (P < 0.05). Compared with ICQG, IL-1β, IL-18, MMP-13 and TNF-α were upregulated in NSAG (P < 0.05), and IL-1β and IL-18 were upregulated in AG (P < 0.05). CONCLUSION The combination of PRP and ICQ can alleviate inflammatory responses in cartilage, promote chondrocyte regeneration and facilitate matrix tissue repair. Compared with single interventions, a synergistic effect is observed.
Collapse
Affiliation(s)
- Liang Cheng
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
- Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- Human Movement Science, Sichuan Sports College, Chengdu, China
| | - Kun Wang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Shuwan Chang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
- Human Movement Science, Sichuan Sports College, Chengdu, China
| | - Yajun Tan
- Affiliated Sport Hospital of Chengdu Sport University, Chengdu, China
| | - Benxiang He
- Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| |
Collapse
|
91
|
Hu Q, Williams SL, Palladino A, Ecker M. Screening of MMP-13 Inhibitors Using a GelMA-Alginate Interpenetrating Network Hydrogel-Based Model Mimicking Cytokine-Induced Key Features of Osteoarthritis In Vitro. Polymers (Basel) 2024; 16:1572. [PMID: 38891518 PMCID: PMC11174780 DOI: 10.3390/polym16111572] [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: 05/01/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by irreversible cartilage degradation. Current clinical treatment options lack effective pharmaceutical interventions targeting the disease's root causes. MMP (matrix metalloproteinase) inhibitors represent a new approach to slowing OA progression by addressing cartilage degradation mechanisms. However, very few drugs within this class are in preclinical or clinical trial phases. Hydrogel-based 3D in vitro models have shown promise as preclinical testing platforms due to their resemblance to native extracellular matrix (ECM), abundant availability, and ease of use. Metalloproteinase-13 (MMP-13) is thought to be a major contributor to the degradation of articular cartilage in OA by aggressively breaking down type II collagen. This study focused on testing MMP-13 inhibitors using a GelMA-alginate hydrogel-based OA model induced by cytokines interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α). The results demonstrate a significant inhibition of type II collagen breakdown by measuring C2C concentration using ELISA after treatment with MMP-13 inhibitors. However, inconsistencies in human cartilage explant samples led to inconclusive results. Nonetheless, the study highlights the GelMA-alginate hydrogel-based OA model as an alternative to human-sourced cartilage explants for in vitro drug screening.
Collapse
Affiliation(s)
- Qichan Hu
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Steven L. Williams
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA
| | - Alessandra Palladino
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| |
Collapse
|
92
|
Yu J, Wang W, Jiang Z, Liu H. TPX2 upregulates MMP13 to promote the progression of lipopolysaccharide-induced osteoarthritis. PeerJ 2024; 12:e17032. [PMID: 38770093 PMCID: PMC11104344 DOI: 10.7717/peerj.17032] [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: 09/07/2023] [Accepted: 02/08/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose This study seeks to identify potential clinical biomarkers for osteoarthritis (OA) using bioinformatics and investigate OA mechanisms through cellular assays. Methods Differentially Expressed Genes (DEGs) from GSE52042 (four OA samples, four control samples) were screened and analyzed with protein-protein interaction (PPI) analysis. Overlapping genes in GSE52042 and GSE206848 (seven OA samples, and seven control samples) were identified and evaluated using Gene Set Enrichment Analysis (GSEA) and clinical diagnostic value analysis to determine the hub gene. Finally, whether and how the hub gene impacts LPS-induced OA progression was explored by in vitro experiments, including Western blotting (WB), co-immunoprecipitation (Co-IP), flow cytometry, etc. Result Bioinformatics analysis of DEGs (142 up-regulated and 171 down-regulated) in GSE52042 identified two overlapping genes (U2AF2, TPX2) that exhibit significant clinical diagnostic value. These genes are up-regulated in OA samples from both GSE52042 and GSE206848 datasets. Notably, TPX2, which AUC = 0.873 was identified as the hub gene. In vitro experiments have demonstrated that silencing TPX2 can alleviate damage to chondrocytes induced by lipopolysaccharide (LPS). Furthermore, there is a protein interaction between TPX2 and MMP13 in OA. Excessive MMP13 can attenuate the effects of TPX2 knockdown on LPS-induced changes in OA protein expression, cell growth, and apoptosis. Conclusion In conclusion, our findings shed light on the molecular mechanisms of OA and suggested TPX2 as a potential therapeutic target. TPX2 could promote the progression of LPS-induced OA by up-regulating the expression of MMP13, which provides some implications for clinical research.
Collapse
Affiliation(s)
- Jingtao Yu
- Department of Orthopedic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Weiqi Wang
- Department of Orthopedic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Zenghui Jiang
- Department of Orthopedic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Huashun Liu
- Department of Orthopedic Surgery, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
93
|
Zhou T, Xiong H, Yao SY, Wang S, Li S, Chang J, Zhai Z, Guo DS, Fan C, Gao C. Hypoxia and Matrix Metalloproteinase 13-Responsive Hydrogel Microspheres Alleviate Osteoarthritis Progression In Vivo. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308599. [PMID: 38054626 DOI: 10.1002/smll.202308599] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/06/2023] [Indexed: 12/07/2023]
Abstract
The occurrence of osteoarthritis (OA) is highly associated with the inflammatory hypoxic microenvironment. Yet currently no attention has been paid to fabricating hypoxia-responsive platforms for OA treatment. Herein, an injectable hydrogel microsphere system (HAM-SA@HCQ) focusing on the hypoxic inflamed joint is prepared with methacrylate-modified sulfonated azocalix[4]arene (SAC4A-MA), methacrylated hyaluronic acid (HA-MA), and dithiol-terminated matrix metalloproteinase 13 (MMP-13) sensitive peptide via a microfluidic device and photo crosslinking technique, followed by encapsulation of the anti-inflammatory drug hydroxychloroquine (HCQ) through host-guest interaction. Owing to the hydrophobic deep cavity, phenolic units, and azo bonds of SAC4A-MA, the hydrogel microspheres show strong drug loading capacity, prominent reactive oxygen species (ROS) scavenging capability, and specific hypoxia-responsive drug release ability. In the OA tissue microenvironment, the hydrogel microspheres undergo degradation by excessive MMP-13 and release HCQ under the hypoxia condition, which synergizes with the ROS-scavenging calixarene to inhibit the inflammatory response of macrophages. After being injected into the OA-inflamed joint, the HAM-SA@HCQ can significantly attenuate the oxidative stress, downregulate the expression of hypoxia-induced factor-1α and inflammatory cytokines, and prevent the cartilage from being destroyed.
Collapse
Affiliation(s)
- Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Hao Xiong
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Shun-Yu Yao
- College of Chemistry, Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shifen Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jieting Chang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
94
|
Antoinette AY, Ziemian SN, Brown AR, Hudson EB, Chlebek C, Wright TM, Goldring SR, Goldring MB, Otero M, van der Meulen MC. PTH treatment before cyclic joint loading improves cartilage health and attenuates load-induced osteoarthritis development in mice. SCIENCE ADVANCES 2024; 10:eadk8402. [PMID: 38640238 PMCID: PMC11029811 DOI: 10.1126/sciadv.adk8402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/18/2024] [Indexed: 04/21/2024]
Abstract
Osteoarthritis (OA) treatment is limited by the lack of effective nonsurgical interventions to slow disease progression. Here, we examined the contributions of the subchondral bone properties to OA development. We used parathyroid hormone (PTH) to modulate bone mass before OA initiation and alendronate (ALN) to inhibit bone remodeling during OA progression. We examined the spatiotemporal progression of joint damage by combining histopathological and transcriptomic analyses across joint tissues. The additive effect of PTH pretreatment before OA initiation and ALN treatment during OA progression most effectively attenuated load-induced OA pathology. Individually, PTH directly improved cartilage health and slowed the development of cartilage damage, whereas ALN primarily attenuated subchondral bone changes associated with OA progression. Joint damage reflected early transcriptomic changes. With both treatments, the structural changes were associated with early modulation of immunoregulation and immunoresponse pathways that may contribute to disease mechanisms. Overall, our results demonstrate the potential of subchondral bone-modifying therapies to slow the progression of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Miguel Otero
- Hospital for Special Surgery, New York, NY, USA
- Weill Cornell Medicine, New York, NY, USA
| | | |
Collapse
|
95
|
Jo HG, Baek CY, Lee J, Hwang Y, Baek E, Hwang JH, Lee D. Anti-Inflammatory, Analgesic, Functional Improvement, and Chondroprotective Effects of Erigeron breviscapus (Vant.) Hand.-Mazz. Extract in Osteoarthritis: An In Vivo and In Vitro Study. Nutrients 2024; 16:1035. [PMID: 38613068 PMCID: PMC11013172 DOI: 10.3390/nu16071035] [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: 03/09/2024] [Revised: 03/31/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative bone disease characterized by inflammation as a primary pathology and currently lacks therapeutic interventions to impede its progression. Erigeron breviscapus (Vant.) Hand.-Mazz. (EB) is an east Asian herbal medicine with a long history of use and a wide range of confirmed efficacy against cardiovascular and central nervous system diseases. The purpose of this study is to evaluate whether EB is worthy of further investigation as a treatment for OA based on anti-inflammatory activity. This study aims to assess the potential of EB as a treatment for OA, focusing on its anti-inflammatory properties. Analgesic effects, functional improvements, and inhibition of cartilage destruction induced by EB were evaluated in acetic acid-induced peripheral pain mice and monosodium iodoacetate-induced OA rat models. Additionally, the anti-inflammatory effect of EB was assessed in serum and cartilage tissue in vivo, as well as in lipopolysaccharide-induced RAW 264.7 cells. EB demonstrated a significant alleviation of pain, functional impairment, and cartilage degradation in OA along with a notable inhibition of pro-inflammatory cytokines, including interleukin-1β, interleukin-6, matrix metalloproteinases 13, and nitric oxide synthase 2, both in vitro and in vivo, in a dose-dependent manner compared to the active control. Accordingly, EB merits further exploration as a potential disease-modifying drug for OA, capable of mitigating the multifaceted pathology of osteoarthritis through its anti-inflammatory properties. Nonetheless, additional validation through a broader experimental design is essential to substantiate the findings of this study.
Collapse
Affiliation(s)
- Hee-Geun Jo
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
- Naturalis Inc., 6, Daewangpangyo-ro, Bundang-gu, Seongnam-si 13549, Republic of Korea
| | - Chae Yun Baek
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| | - JunI Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| | - Yeseul Hwang
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| | - Eunhye Baek
- RexSoft Inc., 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Ji Hye Hwang
- Department of Acupuncture and Moxibustion Medicine, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Donghun Lee
- Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam-si 13120, Republic of Korea; (H.-G.J.)
| |
Collapse
|
96
|
Luo J, Zhou J, Luo JZ, Wang HL, Zhao XL, Zhou RD. Inhibiting MMP13 Attenuates Deep Vein Thrombosis in a Mouse Model by Reducing the Expression of Pdpn. Curr Med Sci 2024; 44:369-379. [PMID: 38619683 DOI: 10.1007/s11596-024-2862-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/28/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVE Matrix metalloproteinase 13 (MMP13) is an extracellular matrix protease that affects the progression of atherosclerotic plaques and arterial thrombi by degrading collagens, modifying protein structures and regulating inflammatory responses, but its role in deep vein thrombosis (DVT) has not been determined. The purpose of this study was to investigate the potential effects of MMP13 and MMP13-related genes on the formation of DVT. METHODS We altered the expression level of MMP13 in vivo and conducted a transcriptome study to examine the expression and relationship between MMP13 and MMP13-related genes in a mouse model of DVT. After screening genes possibly related to MMP13 in DVT mice, the expression levels of candidate genes in human umbilical vein endothelial cells (HUVECs) and the venous wall were evaluated. The effect of MMP13 on platelet aggregation in HUVECs was investigated in vitro. RESULTS Among the differentially expressed genes, interleukin 1 beta, podoplanin (Pdpn), and factor VIII von Willebrand factor (F8VWF) were selected for analysis in mice. When MMP13 was inhibited, the expression level of PDPN decreased significantly in vitro. In HUVECs, overexpression of MMP13 led to an increase in the expression level of PDPN and induced platelet aggregation, while transfection of PDPN-siRNA weakened the ability of MMP13 to increase platelet aggregation. CONCLUSIONS Inhibiting the expression of MMP13 could reduce the burden of DVT in mice. The mechanism involves downregulating the expression of Pdpn through MMP13, which could provide a novel gene target for DVT diagnosis and treatment.
Collapse
Affiliation(s)
- Ji Luo
- Department of Intensive Care Unit, The First People's Hospital of Ziyang, Ziyang, 641300, China
| | - Jin Zhou
- Department of Orthopedics, Kunming Medical University Second Affiliated Hospital, Kunming, 650033, China
| | - Jing-Zeng Luo
- Department of Orthopedics, Kunming Medical University First Affiliated Hospital, Kunming, 650032, China
| | - Hai-Long Wang
- Department of Orthopedics, The Third People's Hospital of Yunnan Province, Kunming, 650200, China
| | - Xue-Ling Zhao
- Department of Orthopedics, Kunming Medical University First Affiliated Hospital, Kunming, 650032, China
| | - Ru-Dan Zhou
- Department of Orthopedics, Kunming Medical University First Affiliated Hospital, Kunming, 650032, China.
| |
Collapse
|
97
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
98
|
Xu JX, Xu FZ, Furbish A, Braxton AM, Brumfield B, Helke KL, Peterson YK. Inhibition of complement C3 prevents osteoarthritis progression in guinea pigs by blocking STAT1 activation. Commun Biol 2024; 7:370. [PMID: 38538870 PMCID: PMC10973449 DOI: 10.1038/s42003-024-06051-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/14/2024] [Indexed: 12/14/2024] Open
Abstract
Osteoarthritis (OA) is one of the leading causes of disability, affecting over 500 million adults worldwide. Previous studies have found that various inflammatory factors can contribute to the pathogenesis of OA, including complement factors in the synovial fluid of OA patients. However, the pathogenesis of this disease is still not known, and the only therapy of severe OA is total joint replacements. Total joint replacements are invasive, expensive, and affect quality of life. Here we show that when human articular chondrocytes are stimulated with pro-inflammatory mediator interleukin-1β (IL-1β) there is an increase in inflammatory factors including complement component 3 (C3). We also found the transcription factor, signal transducer and activator of transcription 1 (STAT1), is responsible for increased C3 expression after IL-1β stimulation in human articular chondrocytes. A specific STAT1 inhibitor, fludarabine, attenuates the hyper-expression of C3 and delays/prevents spontaneous OA in Dunkin-Hartley guinea pigs. Since fludarabine is already clinically used for chemotherapy, this study has great translational potential as a unique disease-modifying osteoarthritis drug (DMOAD) in treating primary OA.
Collapse
Affiliation(s)
- Jen X Xu
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President Street, Charleston, SC, 29425, USA.
| | - Frank Z Xu
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President Street, Charleston, SC, 29425, USA
- UAB Heersink School of Medicine, Alabama, AL, 35233, USA
| | - Amelia Furbish
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President Street, Charleston, SC, 29425, USA
| | - Alicia M Braxton
- Department of Comparative Medicine, Medical University of South Carolina, 114 Doughty Street, Charleston, SC, 29425, USA
| | - Brook Brumfield
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President Street, Charleston, SC, 29425, USA
| | - Kristi L Helke
- Department of Comparative Medicine, Medical University of South Carolina, 114 Doughty Street, Charleston, SC, 29425, USA
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President Street, Charleston, SC, 29425, USA.
| |
Collapse
|
99
|
You X, Ye Y, Lin S, Zhang Z, Guo H, Ye H. Identification of key genes and immune infiltration in osteoarthritis through analysis of zinc metabolism-related genes. BMC Musculoskelet Disord 2024; 25:227. [PMID: 38509535 PMCID: PMC10956297 DOI: 10.1186/s12891-024-07347-8] [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: 10/26/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) represents a prominent etiology of considerable pain and disability, and conventional imaging methods lack sensitivity in diagnosing certain types of OA. Therefore, there is a need to identify highly sensitive and efficient biomarkers for OA diagnosis. Zinc ions feature in the pathogenesis of OA. This work aimed to investugate the role of zinc metabolism-related genes (ZMRGs) in OA and the diagnostic characteristics of key genes. METHODS We obtained datasets GSE169077 and GSE55235 from the Gene Expression Omnibus (GEO) and obtained ZMRGs from MSigDB. Differential expression analysis was conducted on the GSE169077 dataset using the limma R package to identify differentially expressed genes (DEGs), and the intersection of DEGs and ZMRGs yielded zinc metabolism differential expression-related genes (ZMRGs-DEGs). The clusterProfiler R package was employed for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of ZMRGs-DEGs. Potential small molecule drugs were predicted using the CMap database, and immune cell infiltration and function in OA individuals were analyzed using the ssGSEA method. Protein-protein interaction (PPI) networks were constructed to detect Hub genes among ZMRGs-DEGs. Hub gene expression levels were analyzed in the GSE169077 and GSE55235 datasets, and their diagnostic characteristics were assessed using receiver operating characteristic (ROC) curves. The gene-miRNA interaction network of Hub genes was explored using the gene-miRNA interaction network website. RESULTS We identified 842 DEGs in the GSE169077 dataset, and their intersection with ZMRGs resulted in 46 ZMRGs-DEGs. ZMRGs-DEGs were primarily enriched in functions such as collagen catabolic processes, extracellular matrix organization, metallopeptidase activity, and pathways like the IL-17 signaling pathway, Nitrogen metabolism, and Relaxin signaling pathway. Ten potential small-molecule drugs were predicted using the CMap database. OA patients exhibited distinct immune cell abundance and function compared to healthy individuals. We identified 4 Hub genes (MMP2, MMP3, MMP9, MMP13) through the PPI network, which were highly expressed in OA and demonstrated good diagnostic performance. Furthermore, two closely related miRNAs for each of the 4 Hub genes were identified. CONCLUSION 4 Hub genes were identified as potential diagnostic biomarkers and therapeutic targets for OA.
Collapse
Affiliation(s)
- Xiaoxuan You
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, 34 Zhongshan North Road, Licheng District, Quanzhou, 362000, Fujian, China
| | - Yanbo Ye
- Suzhou University Medical Department, Suzhou, 215000, Jiangsu, China
| | - Shufeng Lin
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, 34 Zhongshan North Road, Licheng District, Quanzhou, 362000, Fujian, China
| | - Zefeng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, 34 Zhongshan North Road, Licheng District, Quanzhou, 362000, Fujian, China
| | - Huiyang Guo
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, 34 Zhongshan North Road, Licheng District, Quanzhou, 362000, Fujian, China
| | - Hui Ye
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, 34 Zhongshan North Road, Licheng District, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
100
|
Cui SH, Yan Y, Lu A, Dou Y, Li ZW, Zhu ZH, Du MZ, Zhu YF, Chen X, Wang X, Jiang LX, Shi Y, Liu X, Zhu YJ, Jiang D, Wang JC. Nanomedicines Promote Cartilage Regeneration in Osteoarthritis by Synergistically Enhancing Chondrogenesis of Mesenchymal Stem Cells and Regulating Inflammatory Environment. ACS NANO 2024; 18:8125-8142. [PMID: 38451090 DOI: 10.1021/acsnano.3c11848] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive erosion of the articular cartilage and inflammation. Mesenchymal stem cells' (MSCs) transplantation in OA treatment is emerging, but its clinical application is still limited by the low efficiency in oriented differentiation. In our study, to improve the therapeutic efficiencies of MSCs in OA treatment by carbonic anhydrase IX (CA9) siRNA (siCA9)-based inflammation regulation and Kartogenin (KGN)-based chondrogenic differentiation, the combination strategy of MSCs and the nanomedicine codelivering KGN and siCA9 (AHK-CaP/siCA9 NPs) was used. In vitro results demonstrated that these NPs could improve the inflammatory microenvironment through repolarization of M1 macrophages to the M2 phenotype by downregulating the expression levels of CA9 mRNA. Meanwhile, these NPs could also enhance the chondrogenesis of bone marrow-derived mesenchymal stem cells (BMSCs) by upregulating the pro-chondrogenic TGF-β1, ACAN, and Col2α1 mRNA levels. Moreover, in an advanced OA mouse model, compared with BMSCs alone group, the lower synovitis score and OARSI score were found in the group of BMSCs plus AHK-CaP/siCA9 NPs, suggesting that this combination approach could effectively inhibit synovitis and promote cartilage regeneration in OA progression. Therefore, the synchronization of regulating the inflammatory microenvironment through macrophage reprogramming (CA9 gene silencing) and promoting MSCs oriented differentiation through a chondrogenic agent (KGN) may be a potential strategy to maximize the therapeutic efficiency of MSCs for OA treatment.
Collapse
Affiliation(s)
- Shi-He Cui
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yun Dou
- Department of Sports Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Zhen-Wen Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ze-Hang Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ming-Ze Du
- Department of Sports Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yue-Feng Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xin Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiangyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lin-Xia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yujie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Peking University Ningbo Institute of Marine Medicine, Ningbo, 315832, China
| | - Xiaoyan Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Peking University Ningbo Institute of Marine Medicine, Ningbo, 315832, China
| | - Yuan-Jun Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Peking University Ningbo Institute of Marine Medicine, Ningbo, 315832, China
| | - Dong Jiang
- Department of Sports Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Peking University Ningbo Institute of Marine Medicine, Ningbo, 315832, China
| |
Collapse
|