1
|
Guo Y, Wang P, Hu B, Wang L, Zhang Y, Wang J. Kongensin A targeting PI3K attenuates inflammation-induced osteoarthritis by modulating macrophage polarization and alleviating inflammatory signaling. Int Immunopharmacol 2024; 142:112948. [PMID: 39217884 DOI: 10.1016/j.intimp.2024.112948] [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: 06/25/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
The inflammatory microenvironment, polarization of macrophages towards the M1 phenotype, and consequent matrix degradation and senescence of chondrocytes are primary contributors to the degeneration of knee joint cartilage, further exacerbating the progression of osteoarthritis (OA). Kongensin A (KA) is a recently identified natural plant extract exhibiting anti-necrotic apoptosis and anti-inflammatory properties, but the potential efficacy in alleviating OA remains uncertain. The current research lucubrated the effect of KA on the inflammatory microenvironment and macrophage polarization, as well as its regulatory function in extracellular matrix (ECM) metabolism and chondrocyte senescence. Our findings demonstrated that KA can suppress inflammatory signaling, maintain homeostasis between ECM anabolism and catabolism, and suppress chondrocytes senescence. Further investigation elucidated that the mechanism involves the suppression of the PI3K/AKT/NF-κB axis in chondrocytes under inflammatory conditions. Moreover, KA impeded M1 polarization of macrophages via inhibiting PI3K/AKT/NF-κB axis. Subsequently, we treated chondrocytes with macrophages-derived conditioned medium (CM) and revealed that KA can promote ECM anabolism and alleviate chondrocytes senescence by reprogramming macrophage polarization. Consistent with in vitro experiments, in vivo administration of KA demonstrated alleviated cartilage degeneration and delayed progression of OA. Collectively, through obstructing the PI3K/AKT/NF-κB axis, KA can reprogram macrophage polarization, promote matrix metabolism equilibrium, and alleviate chondrocytes senescence, thereby attenuating the pathology of OA. In conclusion, KA may emerge as a promising therapy for OA.
Collapse
Affiliation(s)
- Yuhui Guo
- Department of Orthopaedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China; Department of Orthopaedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Peng Wang
- Department of Orthopaedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China; Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Ling Wang
- Department of Orthopaedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China; Department of Orthopaedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China.
| | - Yingze Zhang
- Department of Orthopaedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China; Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Juan Wang
- Department of Orthopaedic Research Center, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China.
| |
Collapse
|
2
|
Rasmussen S, Skjoldemose E, Jørgensen NK. Intraarticular gold microparticles using hyaluronic acid as the carrier for hip osteoarthritis. A 2-year follow-up pilot study. Sci Rep 2024; 14:26249. [PMID: 39482349 PMCID: PMC11527870 DOI: 10.1038/s41598-024-77760-5] [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: 04/14/2024] [Accepted: 10/24/2024] [Indexed: 11/03/2024] Open
Abstract
We recently conducted a pilot study in which we discovered that introducing metallic gold microparticles into the knee joint can help reduce pain caused by osteoarthritis for up to two years. Additionally, we identified significant proteomic changes in the synovial fluid and serum within eight weeks of the procedure. In this study, we aimed to evaluate whether there may be a clinical relevant effect of intra-articular injection of gold microparticles on hip osteoarthritis. A cohort of 22 patients, aged ≥ 18 years, with pain ≥ 3 months, and Kellgren-Lawrence OA grade 2-4, were included. Metallic gold 20 mg, 72.000 pieces, 20-40 µ-meter BerlockMicroImplants (BMI) were injected into the hip joint using hyaluronic acid as the carrier. In total, we treated 26 hip joints. The primary outcome was the Western Ontario and McMaster Universities Arthritis Index (WOMAC). WOMAC pain decreased from 11 (2-20 to 3 (0-8), stiffness from 6 (0-8) to 1 (0-4), and activity from 43 (18-68) to 11 (0-27), all P = 0.0001. When adjusting for the minimally relevant differences, the P-values were 0.0015 for WOMAC pain, 0.26 for stiffness, and 0.011 for activity. Combined intraarticular treatment with metallic gold microparticles and hyaluronic acid may improve hip joint pain and function. Joint stiffness did not improve when assessed against the minimal clinically relevant difference. This study suggests a basis for a future placebo-controlled randomized trial of gold microparticles and hyaluronic acid in hip osteoarthritic patients.
Collapse
Affiliation(s)
- Sten Rasmussen
- Department of Clinical Medicine, Aalborg University, 249 Selma Lagerløfs Vej, room 11.03.025, DK-9260, Aalborg, Gistrup, Denmark.
- Department of orthopedic surgery, Aalborg University Hospital, Aalborg, Denmark.
| | - Emilie Skjoldemose
- Department of Clinical Medicine, Aalborg University, 249 Selma Lagerløfs Vej, room 11.03.025, DK-9260, Aalborg, Gistrup, Denmark
| | - Nia Kristine Jørgensen
- Department of Clinical Medicine, Aalborg University, 249 Selma Lagerløfs Vej, room 11.03.025, DK-9260, Aalborg, Gistrup, Denmark
| |
Collapse
|
3
|
Fu W, Fang Y, Wang T, Lu Q, Wu J, Yang Q. Low-Protein Diet Inhibits the Synovial Tissue Macrophage Pro-Inflammatory Polarization Via NRF2/SIRT3/SOD2/ROS Pathway in K/BxN Rheumatoid Arthritis Mice. Inflammation 2024:10.1007/s10753-024-02145-9. [PMID: 39292325 DOI: 10.1007/s10753-024-02145-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/24/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disorder characterized by pain, swelling, stiffness, and impaired function. Attenuating inflammation is a crucial objective in RA management. Diet and nutrition are believed to influence RA symptomatology, with a low-protein diet being one potential nutritional strategy, although its underlying mechanisms remain to be fully elucidated. In this research, serum derived from arthritic transgenic K/BxN mice was administered to naive mice to establish a K/BxN rheumatoid arthritis model. Physiological assessments and histological staining were performed to evaluate joint pathology. (Enzyme-linked immunosorbent assay) ELISA was used to measure inflammatory cytokines. Flow cytometry and immunofluorescence were applied to characterize macrophage phenotypes. Transcriptomic analysis elucidated molecular pathways under the effect of a low-protein diet and verified by immunoblotting. Mitochondrial reactive oxygen species (ROS) was detected by Mito-SOX. Protein expression was silenced through the application of siRNA transfection. Our results indicate that a low-protein diet significantly alleviates disease symptoms and decreases pro-inflammatory cytokine levels in synovial fluid. Furthermore, this dietary intervention inhibits M1 macrophage polarization while promoting a shift towards the M2 phenotype. Transcriptomic analysis revealed that the beneficial effects of the low-protein diet in alleviating rheumatoid arthritis are closely linked to the NRF2 pathway. In vitro, low protein treatment can promote the activity of NRF2 via inhibiting the ubiquitin mediated proteolysis and activate the NRF2/SIRT3/SOD2 pathway to inhibit the production of ROS, which will further inhibit the M1 macrophage polarization. NRF2 knockdown can abolish the effects of low-protein treatment, indicating that the inhibition of M1 polarization and the anti-inflammatory response induced by low-protein treatment are dependent on NRF2. In summary, our findings propose that low-protein diet can inhibit synovial macrophage M1 polarization via activating NRF2/SIRT3/SOD2 pathway to reduce mitochondrial ROS production. This mechanism effectively decreases synovial inflammation and alleviates RA symptoms.
Collapse
Affiliation(s)
- Weicong Fu
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine: Jinhua Municipal Central Hospital, No.365, Renmin East Road, Wucheng District, Jinhua City, 321000, Zhejiang Province, China
| | - Yinfei Fang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine: Jinhua Municipal Central Hospital Hospital, Jinhua, 321000, Zhejiang, China
| | - Tianbao Wang
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine: Jinhua Municipal Central Hospital, No.365, Renmin East Road, Wucheng District, Jinhua City, 321000, Zhejiang Province, China
| | - Qinglin Lu
- Department of Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine: Jinhua Municipal Central Hospital, Jinhua, 321000, Zhejiang, China
| | - Junqi Wu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine: Jinhua Municipal Central Hospital Hospital, Jinhua, 321000, Zhejiang, China
| | - Qining Yang
- Department of Orthopedics, Affiliated Jinhua Hospital, Zhejiang University School of Medicine: Jinhua Municipal Central Hospital, No.365, Renmin East Road, Wucheng District, Jinhua City, 321000, Zhejiang Province, China.
- Zhejiang University School of Medicine, Hangzhou, 31009, Zhejiang, China.
| |
Collapse
|
4
|
Roseti L, Borciani G, Amore E, Grigolo B. Cannabinoids in the Inflamed Synovium Can Be a Target for the Treatment of Rheumatic Diseases. Int J Mol Sci 2024; 25:9356. [PMID: 39273304 PMCID: PMC11394920 DOI: 10.3390/ijms25179356] [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: 07/18/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
The management of rheumatic diseases has noticeably changed in recent years with the development of targeted therapeutic agents, namely, biological disease-modifying antirheumatic drugs. Identifying essential signaling pathways and factors crucial for the development and progression of these diseases remains a significant challenge. Therapy could be used to delay the onset or reduce harm. The endocannabinoid system's presence within the synovium can be identified as a suggested target for therapeutic interventions due to its role in modulating pain, inflammation, and joint metabolism. This review brings together the most pertinent information concerning the actions of the endocannabinoid system present in inflamed synovial tissue and its interaction with phytocannabinoids and synthetic cannabinoids, which can be used from a therapeutic perspective to minimize the inflammatory and pain processes typical of osteoarthritis and rheumatoid arthritis.
Collapse
Affiliation(s)
- Livia Roseti
- RAMSES Laboratory, Rizzoli RIT-Research, Innovation & Technology Department, Istituto di Ricerca Codivilla Putti, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Giorgia Borciani
- RAMSES Laboratory, Rizzoli RIT-Research, Innovation & Technology Department, Istituto di Ricerca Codivilla Putti, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Emanuela Amore
- RAMSES Laboratory, Rizzoli RIT-Research, Innovation & Technology Department, Istituto di Ricerca Codivilla Putti, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Brunella Grigolo
- RAMSES Laboratory, Rizzoli RIT-Research, Innovation & Technology Department, Istituto di Ricerca Codivilla Putti, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| |
Collapse
|
5
|
DiNicola ES, Martinez AV, Walker L, Wu Y, Burnikel BG, Mercuri J. Cigarette smoke extract exacerbates progression of osteoarthritic-like changes in cartilage explant cultures. J Orthop Res 2024; 42:1682-1695. [PMID: 38460961 DOI: 10.1002/jor.25828] [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: 11/16/2023] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 03/11/2024]
Abstract
Established risk factors for osteoarthritis (OA) include obesity, joint injury, age, race, and genetics. However, the relationship between cigarette smoking and OA has yet to be established. In the present study, we have employed the use of cigarette smoke extract (CSE), the water-soluble vapor phase of cigarette smoke, with porcine cartilage explants to investigate the effects of cigarette smoking on cartilage catabolism at the tissue level. Articular cartilage explants were first exposed to 2.5%, 5%, and 10% CSE to assess its effects on cartilage homeostasis. Following, the effects of CSE on OA-like inflammation was observed by culturing explants with a combined treatment of IL-1β and TNF-α and 10% CSE (CSE + OA). Cartilage explants were assessed for changes in viability, biochemical composition, extracellular matrix (ECM) integrity, and equilibrium mechanical properties (aggregate modulus and hydraulic permeability). CSE alone leads to both a time- and dose-dependent decrease in chondrocyte viability but does not significantly affect sGAG content, percent sGAG loss, or the ECM integrity of cartilage explants. When IL-1β and TNF-α were combined with 10% CSE, this led to a synergistic effect with more significant losses in viability, significantly more sGAG loss, and significantly higher production of ROS than OA-like inflammation only. Cartilage explant equilibrium mechanical properties were unaffected. Within the timeframe of this study, CSE alone does not cause OA but when combined with OA-like inflammation leads to worsened articular cartilage degeneration as measured by chondrocyte viability, sGAG loss, proteoglycan staining, and ROS production.
Collapse
Affiliation(s)
- Emily Sawvell DiNicola
- Department of Bioengineering, The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, USA
- Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, South Carolina, USA
| | - Andrea Vera Martinez
- Department of Bioengineering, The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, USA
- Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, South Carolina, USA
| | - Lizzie Walker
- Orthopaedic Bioengineering Laboratory, Medical University of South Carolina, Department of Bioengineering, Clemson University, Charleston, South Carolina, USA
| | - Yongren Wu
- Orthopaedic Bioengineering Laboratory, Medical University of South Carolina, Department of Bioengineering, Clemson University, Charleston, South Carolina, USA
| | - Brian G Burnikel
- Prisma Health Steadman Hawkins Clinic of the Carolinas - Patewood, Greenville, South Carolina, USA
| | - Jeremy Mercuri
- Department of Bioengineering, The Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, USA
- Frank H. Stelling and C. Dayton Riddle Orthopaedic Education and Research Laboratory, Clemson University Biomedical Engineering Innovation Campus, Greenville, South Carolina, USA
| |
Collapse
|
6
|
Nayak AG, Bhatnagar S, Pai Khot AJ. Correlation Between Radiological Changes of the Temporomandibular Joint and Upper Cervical Vertebrae in Degenerative Joint Disease: A Cone-Beam Computed Tomography-Based Analytical Study. Cureus 2024; 16:e67518. [PMID: 39310450 PMCID: PMC11416149 DOI: 10.7759/cureus.67518] [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] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Objectives This study was conducted to assess the radiological changes of the temporomandibular joint (TMJ) and cervical vertebrae individually and their correlation in degenerative joint disease (DJD) using a cone-beam computed tomography (CBCT)-based approach. Methodology The study employed a cross-sectional, analytical retrospective design, analyzing one-year data. CBCT scans of 60 patients (120 TMJs) were assessed for degenerative changes using standardized imaging parameters. Eligibility criteria included full field-of-view CBCT scans, excluding those with craniofacial anomalies or prior orthodontic treatment. Radiological assessments of TMJs and cervical vertebrae were conducted by experienced radiologists using the Anjos Pontual method and novel grading system (TMJ Spine Degenerative Severity Index). Results The study included 60 CBCT scans (120 joints), with 31.7% males and 68.3% females. Participants were predominantly aged 31-60 years (58.3%). DJD findings for the right TMJ showed grade 1 changes in 55.0% and grade 2 in 31.7%, while the left TMJ had 46.7% grade 1 and 35.0% grade 2 changes. A strong positive correlation (0.704) was found between bilateral TMJ and cervical vertebrae changes. Age correlated significantly with TMJ alterations but not with cervical vertebrae changes. Conclusion This study demonstrated that there exists a positive association between the radiological changes of TMJ and cervical vertebrae in DJD with age, which can be detected in mild stage of severity on CBCT and can be of use in clinical correlation and application of optimal interventions ensuring better prognosis.
Collapse
Affiliation(s)
- Ajay G Nayak
- Department of Maxillofacial Imaging, INsight CBCT (Cone-Beam Computed Tomography) Imaging Center, Mumbai, IND
| | - Sunanda Bhatnagar
- Department of Oral Medicine and Radiology, Terna Public Charitable Trust's Terna Dental College, Navi Mumbai, IND
| | - Atrey J Pai Khot
- Department of Public Health Dentistry, Goa Dental College and Hospital, Bambolim, IND
| |
Collapse
|
7
|
Shen S, Liang Y, Zhao Y, Hu Z, Huang Y, Wu Y, Liu Y, Fan S, Wang Q, Xiao P. Dietary supplementation of vitamin B1 prevents the pathogenesis of osteoarthritis. Proc Natl Acad Sci U S A 2024; 121:e2408160121. [PMID: 39024114 PMCID: PMC11287274 DOI: 10.1073/pnas.2408160121] [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: 04/23/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
As the primary cause for chronic pain and disability in elderly individuals, osteoarthritis (OA) is one of the fastest-growing diseases due to the aging world population. To date, the impact of microenvironmental changes on the pathogenesis of OA remains poorly understood, greatly hindering the development of effective therapeutic approaches against OA. In this study, we profiled the differential metabolites in the synovial fluid from OA patients and identified the downregulation of vitamin B1 (VB1) as a metabolic feature in the OA microenvironment. In a murine destabilization of medial meniscus-induced OA model, supplementation of VB1 significantly mitigated the symptoms of OA. Cytokine array analysis revealed that VB1 treatment remarkably reduced the production of a pro-OA factor-C-C Motif Chemokine Ligand 2 (CCL2), in macrophages. Further evidence demonstrated that exogenous CCL2 counteracted the anti-OA function of VB1. Hence, our study unveils a unique biological function of VB1 and provides promising clues for the diet-based treatment of OA.
Collapse
Affiliation(s)
- Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yi Liang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yuening Zhao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Ziang Hu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Youling Huang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yufei Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu 322000, China
| |
Collapse
|
8
|
Yi X, Leng P, Wang S, Liu L, Xie B. Functional Nanomaterials for the Treatment of Osteoarthritis. Int J Nanomedicine 2024; 19:6731-6756. [PMID: 38979531 PMCID: PMC11230134 DOI: 10.2147/ijn.s465243] [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: 02/21/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, affecting more than 595 million people worldwide. Nanomaterials possess superior physicochemical properties and can influence pathological processes due to their unique structural features, such as size, surface interface, and photoelectromagnetic thermal effects. Unlike traditional OA treatments, which suffer from short half-life, low stability, poor bioavailability, and high systemic toxicity, nanotherapeutic strategies for OA offer longer half-life, enhanced targeting, improved bioavailability, and reduced systemic toxicity. These advantages effectively address the limitations of traditional therapies. This review aims to inspire researchers to develop more multifunctional nanomaterials and promote their practical application in OA treatment.
Collapse
Affiliation(s)
- Xinyue Yi
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Pengyuan Leng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Supeng Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, People’s Republic of China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| | - Bingju Xie
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
9
|
Syed NH, Misbah I, Azlan M, Ahmad Mohd Zain MR, Nurul AA. Exosomes in Osteoarthritis: A Review on Their Isolation Techniques and Therapeutic Potential. Indian J Orthop 2024; 58:866-875. [PMID: 38948378 PMCID: PMC11208382 DOI: 10.1007/s43465-024-01175-7] [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: 03/11/2024] [Accepted: 04/30/2024] [Indexed: 07/02/2024]
Abstract
Background Exosomes are the smallest extracellular vesicles (30-150 nm) secreted by all cell types, including synovial fluid. However, because biological fluids are complex, heterogeneous, and contain contaminants, their isolation is difficult and time-consuming. Furthermore, the pathophysiology of osteoarthritis (OA) involves exosomes carrying complex components that cause macrophages to release chemokines and proinflammatory cytokines. This narrative review aims to provide in-depth insights into exosome biology, isolation techniques, role in OA pathophysiology, and potential role in future OA therapeutics. Methods A literature search was conducted using PubMed, Scopus, and Web of Science databases for studies involving exosomes in the osteoarthritis using keywords "Exosomes" and "Osteoarthritis". Relevant articles in the last 15 years involving both human and animal models were included. Studies involving exosomes in other inflammatory diseases were excluded. Results Despite some progress, conventional techniques for isolating exosomes remain laborious and difficult, requiring intricate and time-consuming procedures across various body fluids and sample origins. Moreover, exosomes are involved in various physiological processes associated with OA, like cartilage calcification, degradation of osteoarthritic joints, and inflammation. Conclusion The process of achieving standardization, integration, and high throughput of exosome isolation equipment is challenging and time-consuming. The integration of various methodologies can be employed to effectively address specific issues by leveraging their complementary benefits. Exosomes have the potential to effectively repair damaged cartilage OA, reduce inflammation, and maintain a balance between the formation and breakdown of cartilage matrix, therefore showing promise as a therapeutic option for OA.
Collapse
Affiliation(s)
- Nazmul Huda Syed
- Center for Global Health Research, Saveetha Medical Collage and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Iffath Misbah
- Department of Radio Diagnosis, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Maryam Azlan
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | | | - Asma Abdullah Nurul
- School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
10
|
Zhang Y, Li Z, Chen C, Wei W, Li Z, Zhou H, He W, Xia J, Li B, Yang Y. SRGN promotes macrophage recruitment through CCL3 in osteoarthritis. Connect Tissue Res 2024; 65:330-342. [PMID: 39067006 DOI: 10.1080/03008207.2024.2380313] [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: 09/08/2023] [Revised: 03/18/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative disease that affects synovial joints and leads to significant pain and disability, particularly in older adults. Infiltration of macrophages plays a key role in the progression of OA. However, the mechanisms underlying macrophage recruitment in OA are not fully understood. METHODS The Serglycin (SRGN) expression pattern was analyzed, along with its association with macrophage infiltration in OA, using bioinformatic methods. SRGN expression in chondrocytes was altered by small interfering RNA (siRNA) and plasmids. Conditioned media (CM) was obtained from transfected chondrocytes to establish a co-culture model of chondrocytes and THP-1 derived macrophages. The impact of SRGN on macrophage recruitment was evaluated using a transwell assay. Furthermore, the regulatory effect of SRGN on CCL3 was validated through qPCR, WB, and ELISA experiments. RESULTS In OA patients, the upregulation of SRGN positively correlated with K-L grade and macrophage infiltration. It was found that SRGN expression and secretion were up-regulated in OA and that it can promote macrophage migration in vitro. Further investigation showed that SRGN affects macrophage migration by regulating the expression of CCL3. CONCLUSION SRGN in chondrocytes plays a role in promoting the recruitment of THP-1 derived macrophages in vitro by regulating production of CCL3.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihua Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Wei
- The First Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhendong Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haichao Zhou
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenbao He
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang Xia
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunfeng Yang
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Luo H, Li L, Han S, Liu T. The role of monocyte/macrophage chemokines in pathogenesis of osteoarthritis: A review. Int J Immunogenet 2024; 51:130-142. [PMID: 38462560 DOI: 10.1111/iji.12664] [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: 12/02/2023] [Revised: 02/08/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024]
Abstract
Osteoarthritis (OA) is one of the most common degenerative diseases characterised by joint pain, swelling and decreased mobility, with its main pathological features being articular synovitis, cartilage degeneration and osteophyte formation. Inflammatory cytokines and chemokines secreted by activated immunocytes can trigger various inflammatory and immune responses in articular cartilage and synovium, contributing to the genesis and development of OA. A series of monocyte/macrophage chemokines, including monocyte chemotaxis protein (MCP)-1/CCL2, MCP2/CCL8, macrophage inflammatory protein (MIP)-1α/CCL3, MIP-1β/CCL4, MIP-3α/CCL20, regulated upon activation, normal T-cell expressed and secreted /CCL5, CCL17 and macrophage-derived chemokine/CCL22, was proven to transmit cell signals by binding to G protein-coupled receptors on recipient cell surface, mediating and promoting inflammation in OA joints. However, the underlying mechanism of these chemokines in the pathogenesis of OA remains still elusive. Here, published literature was reviewed, and the function and mechanisms of monocyte/macrophage chemokines in OA pathogenesis were summarised. The symptoms and disease progression of OA were found to be effectively alleviated when the expression of these chemokines is inhibited. Elucidating these mechanisms could contribute to further understand how OA develops and provide potential targets for the early diagnosis of arthritis and drug treatment to delay or even halt OA progression.
Collapse
Affiliation(s)
- Hao Luo
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Linfeng Li
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Song Han
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Liu
- Department of Orthopaedics, The People's Hospital Affiliated to Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
12
|
Yuan Q, Yang M, Zheng H, Cai Y, Luo P, Wang X, Xu P. M2 Macrophage-Derived Extracellular Vesicles Encapsulated in Hyaluronic Acid Alleviate Osteoarthritis by Modulating Macrophage Polarization. ACS Biomater Sci Eng 2024; 10:3355-3377. [PMID: 38563817 DOI: 10.1021/acsbiomaterials.3c01833] [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: 04/04/2024]
Abstract
An imbalance between M1 and M2 macrophage polarization is critical in osteoarthritis (OA) development. We investigated the effect of M2 macrophage-derived extracellular vesicles (M2-EVs) to reprogramme macrophages from the M1 to M2 phenotype for OA treatment. M1 macrophages and mouse OA models were treated with M2-EVs. Proteomic analysis was performed to evaluate macrophage polarization in vitro. The OA models were as follows: destabilization of the medial meniscus (DMM) surgery-induced OA and collagenase-induced OA (CIOA). Hyaluronic acid (HA) was used to deliver M2-EVs. M2-EVs decreased macrophage accumulation, repolarized macrophages from the M1 to M2 phenotype, mitigated synovitis, reduced cartilage degradation, alleviated subchondral bone damage, and improved gait abnormalities in the CIOA and DMM models. Moreover, HA increased the retention time of M2-EVs and enhanced the efficiency of M2-EVs in OA treatment. Furthermore, proteomic analysis demonstrated that M2-EVs exhibited a macrophage reprogramming ability similar to IL-4, and the pathways might be the NOD-like receptor (NLR), TNF, NF-κB, and Toll-like receptor (TLR) signaling pathways. M2-EVs reprogrammed macrophages from the M1 to M2 phenotype, which resulted in beneficial effects on cartilage and attenuation of OA severity. In summary, our study indicated that M2-EV-guided reprogramming of macrophages is a promising treatment strategy for OA.
Collapse
Affiliation(s)
- Qiling Yuan
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Haishi Zheng
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Yongsong Cai
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Pan Luo
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Xinyi Wang
- Department of Rehabilitation, Shaanxi Provincial Rehabilitation Hospital, Xi'an, Shaanxi 710065, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| |
Collapse
|
13
|
Huang L, Yao Y, Ruan Z, Zhang S, Feng X, Lu C, Zhao J, Yin F, Cao C, Zheng L. Baicalin nanodelivery system based on functionalized metal-organic framework for targeted therapy of osteoarthritis by modulating macrophage polarization. J Nanobiotechnology 2024; 22:221. [PMID: 38724958 PMCID: PMC11080297 DOI: 10.1186/s12951-024-02494-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Intra-articular drugs used to treat osteoarthritis (OA) often suffer from poor pharmacokinetics and stability. Nano-platforms as drug delivery systems for drug delivery are promising for OA therapy. In this study, we reported an M1 macrophage-targeted delivery system Bai@FA-UIO-66-NH2 based on folic acid (FA) -modified metal-organic framework (MOF) loaded with baicalin (Bai) as antioxidant agent for OA therapy. With outstanding biocompatibility and high drug loading efficiency, Bai@FA-UIO-66-NH2 could be specifically uptaken by LPS-induced macrophages to serve as a potent ROS scavenger, gradually releasing Bai at the subcellular level to reduce ROS production, modulate macrophage polarization to M2, leading to alleviation of synovial inflammation in OA joints. The synergistic effect of Bai@FA-UIO-66-NH2 on macrophage polarization and ROS scavenging significantly improved the therapeutic efficacy of OA, which may provide a new insight into the design of OA precision therapy.
Collapse
Affiliation(s)
- Lanli Huang
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Yi Yao
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Zhuren Ruan
- Department of Dermatology and Venereology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shengqing Zhang
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Xianjing Feng
- Pharmaceutical College, Guangxi Medical University, Nanning, 530021, China
| | - Chun Lu
- School of Materials and Environment, Guangxi Minzu University, Nanning, 53000, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China
| | - Feiying Yin
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| | - Cunwei Cao
- Department of Dermatology and Venereology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed By the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
14
|
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: 0] [Impact Index Per Article: 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
|
15
|
Liu T, Li X, Pang M, Wang L, Li Y, Sun X. Machine learning-based endoplasmic reticulum-related diagnostic biomarker and immune microenvironment landscape for osteoarthritis. Aging (Albany NY) 2024; 16:4563-4578. [PMID: 38428406 PMCID: PMC10968715 DOI: 10.18632/aging.205611] [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: 10/13/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease worldwide. Further improving the current limited understanding of osteoarthritis has positive clinical value. METHODS OA samples were collected from GEO database and endoplasmic reticulum related genes (ERRGs) were identified. The WGCNA network was further built to identify the crucial gene module. Based on the expression profiles of characteristic ERRGs, LASSO algorithm was used to select key factors according to the minimum λ value. Random forest (RF) algorithm was used to calculate the importance of ERRGs. Subsequently, overlapping genes based on LASSO and RF algorithms were identified as ERRGs-related diagnostic biomarkers. In addition, OA specimens were also collected and performed qRT-PCR quantitative analysis of selected ERRGs. RESULTS We identified four ERRGs associated with OA risk assessment through machine learning methods, and verified the abnormal expressions of these screened markers in OA patients through in vitro experiments. The influence of selected markers on OA immune infiltration was also evaluated. CONCLUSIONS Our results provide new evidence for the role of ER stress in the OA progression, as well as new markers and potential intervention targets for OA.
Collapse
Affiliation(s)
- Tingting Liu
- Research Center for Drug Safety Evaluation of Hainan, Hainan Medical University, Haikou, Hainan 571199, China
| | - Xiaomao Li
- Jiangsu Food and Pharmaceutical Science College, Huaian, Jiangsu 223023, China
| | - Mu Pang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Shenzhen, Guangdong 518000, China
| | - Lifen Wang
- Research Center for Drug Safety Evaluation of Hainan, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ye Li
- Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - Xizhe Sun
- Research Center for Drug Safety Evaluation of Hainan, Hainan Medical University, Haikou, Hainan 571199, China
| |
Collapse
|
16
|
Zheng S, Li Y, Yin L, Lu M. Identification of sulfur metabolism-related gene signature in osteoarthritis and TM9SF2's sustenance effect on M2 macrophages' phagocytic activity. J Orthop Surg Res 2024; 19:62. [PMID: 38218914 PMCID: PMC10787471 DOI: 10.1186/s13018-023-04384-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/18/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic and low-grade inflammatory disease associated with metabolism disorder and multiple cell death types in the synovial tissues. Sulfur metabolism has not been studied in OA. METHODS First, we calculated the single sample gene set enrichment analysis score of sulfur metabolism-associated annotations (i.e., cysteine metabolism process, regulation of sulfur metabolism process, and disulfidptosis) between healthy and synovial samples from patients with OA. Sulfur metabolism-related differentially expressed genes (DEGs) were analyzed in OA. Least absolute shrinkage and selection operator COX regression were used to identify the sulfur metabolism-associated gene signature for diagnosing OA. Correlation and immune cell deconvolution analyses were used to explore the correlated functions and cell specificity of the signature gene, TM9SF2. TM9SF2's effect on the phagocytosis of macrophages M2 was analyzed by coculturing macrophages with IgG-coated beads or apoptotic Jurkat cells. RESULTS A diagnostic six gene signature (i.e., MTHFD1, PDK4, TM9SF2, POU4F1, HOXA2, NCKAP1) was identified based on the ten DEGs, validated using GSE12021 and GSE1919 datasets. TM9SF2 was upregulated in the synovial tissues of OA at both mRNA and protein levels. The relationship between TM9SF2 and several functional annotations, such as antigen processing and presentation, lysosome, phagosome, Fcγ-mediated phagocytosis, and tyrosine metabolism, was identified. TM9SF2 and macrophages M2 were significantly correlated. After silencing TM9SF2 in THP-1-derived macrophages M2, a significantly reduced phagocytosis and attenuated activation of PLC-γ1 were observed. CONCLUSION A sulfur metabolism-associated six-gene signature for OA diagnosis was constructed and upregulation of the phagocytosis-associated gene, TM9SF2, was identified. The findings are expected to deepen our understanding of the molecular mechanism underlying OA development and be used as potential therapeutic targets.
Collapse
Affiliation(s)
- Shuang Zheng
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China.
| | - Yetian Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China
| | - Li Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China
| | - Ming Lu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, No.218 Ji Xi Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
17
|
Jeon S, Min Kim T, Kwon G, Park J, Park SY, Lee SH, Jin EJ. Targeting ROS in osteoclasts within the OA environment: A novel therapeutic strategy for osteoarthritis management. J Tissue Eng 2024; 15:20417314241279935. [PMID: 39483974 PMCID: PMC11526208 DOI: 10.1177/20417314241279935] [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] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/16/2024] [Indexed: 11/03/2024] Open
Abstract
This study investigated the therapeutic potential of a manganese dioxide-polymer dot (MnO2-PD)-incorporated hydrogel, designated as M-PD hydrogel, for modulating reactive oxygen species (ROS) within the osteoarthritis (OA) environment. Our research highlights the ability of the hydrogel to scavenge ROS, thereby influencing the differentiation of osteoclasts and protecting chondrocytes, offering a novel approach to osteoarthritis (OA) management. Our results indicated that the M-PD hydrogel increased electrical resistance and fluorescence recovery in the presence of osteoclasts, correlating with decreased ROS levels and suppressed expression of osteoclast differentiation markers. Coculture experiments revealed the protective effects of the hydrogel on chondrocytes by reducing the expression of matrix-degrading enzymes. In vivo application in burr holes and/or OA-induced mice revealed a significant reduction in osteoclast formation and cartilage destruction, suggesting the dual therapeutic action of the hydrogel in altering the joint microenvironment. These findings highlight the potential of targeting ROS in osteoclasts as a comprehensive therapeutic approach, offering not only symptomatic relief but also targeting the underlying mechanisms of disease progression in OA.
Collapse
Affiliation(s)
- Seungho Jeon
- Department of Biological Sciences, Wonkwang, University, Iksan, Jeonbuk, South Korea
| | - Tae Min Kim
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, South Korea
| | - Gitae Kwon
- Department of Biological Sciences, Wonkwang, University, Iksan, Jeonbuk, South Korea
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan, Jeonbuk, Korea
| | - Junyoung Park
- Department of Biological Sciences, Wonkwang, University, Iksan, Jeonbuk, South Korea
| | - Sung Young Park
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, South Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, South Korea
| | - Seoung Hoon Lee
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan, Jeonbuk, Korea
- Integrated Omics Institute, Wonkwang University, Iksan, Jeonbuk, South Korea
| | - Eun-Jung Jin
- Department of Biological Sciences, Wonkwang, University, Iksan, Jeonbuk, South Korea
- Integrated Omics Institute, Wonkwang University, Iksan, Jeonbuk, South Korea
| |
Collapse
|
18
|
Zhang Y, Ji Q. Macrophage polarization in osteoarthritis progression: a promising therapeutic target. Front Cell Dev Biol 2023; 11:1269724. [PMID: 37954210 PMCID: PMC10639142 DOI: 10.3389/fcell.2023.1269724] [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: 07/30/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Osteoarthritis (OA) is one of the leading causes of pain and disability in the elderly. Synovitis, cartilage destruction and osteophyte formation histologically manifest OA. Unfortunately, there is currently no effective therapy to delay its progression and the underlying mechanisms of OA require further exploration. Macrophage is a main cellular component of joint synovium. It is highly plastic and can be stimulated to polarize to different phenotypes, namely, the pro-inflammatory phenotype (M1) and the anti-inflammatory/tissue-repairing phenotype (M2). Ample evidence has demonstrated the vital roles of macrophages in the progression of OA. Imbalanced M1/M2 ratio is significantly related to OA severity indicating macrophage polarization might be a promising therapeutic target for OA. In this review, we summarized the involvements of polarized macrophages in synovitis, cartilage degradation, osteophyte formation and OA-related chronic pain. Promising therapies targeting macrophage polarization including the intra-articular cell/derivates-based therapy and the alternative non-invasive intervention such as photobiomodulation therapy were reviewed as well.
Collapse
Affiliation(s)
| | - Quanbo Ji
- Department of Orthopedics, The General Hospital of Chinese PLA, Beijing, China
| |
Collapse
|
19
|
Chen GY, Liu XY, Yan XE, Yu X, Liu Y, Luo J, Tao QW. Total Flavonoids of Rhizoma Drynariae Treat Osteoarthritis by Inhibiting Arachidonic Acid Metabolites Through AMPK/NFκB Pathway. J Inflamm Res 2023; 16:4123-4140. [PMID: 37750171 PMCID: PMC10518150 DOI: 10.2147/jir.s418345] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 09/02/2023] [Indexed: 09/27/2023] Open
Abstract
Objective Previous clinical studies have found that total flavonoids of Rhizoma Drynariae (TFRD) have a good therapeutic effect on osteoarthritis (OA), but its therapeutic mechanism needs further research. Methods OA rat model was established by Hulth method and was intervened by TFRD. Pathological assessments were conducted to assess the protective effect of TFRD on cartilage. Serum metabolomics and network pharmacology were detected to predict the mechanism of TFRD treating OA. In further experiments, molecular biology experiment was carried out to confirm the predicted mechanisms in vivo and in vitro. Results TFRD can effectively reduce chondrocyte apoptosis and cartilage degeneration in OA model rats. Serum metabolomics revealed that the intervention effect may be closely related to arachidonic acid metabolism pathway. Network pharmacologic prediction showed that COX-2 was the key target of TFRD in treating OA, and its mechanism might be related with NFκB, apoptosis, AMPK and arachidonic acid metabolism pathway. In vivo experiments indicated that TFRD can inhibit the abnormal expression of COX-2 mRNA in OA model rats. In the in vitro studies, the expression of COX-2 mRNA and protein increased, AMPK phosphorylation was inhibited, and NFκB signaling pathway was activated in IL-1β-induced chondrocytes, and these changes can be reversed by TFRD. After the activation of AMPK signaling pathway or the block-down of NFκB signaling pathway, the effect of TFRD on COX-2 mRNA expression was significantly weakened. Conclusion TFRD can inhibit COX-2-mediated arachidonic acid metabolites, and its mechanism is closely related to AMPK/NFκB pathway, which may be a key mechanism in the treatment of OA.
Collapse
Affiliation(s)
- Guang-Yao Chen
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Xiao-Yu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Xue-Er Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - XinBo Yu
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yi Liu
- Humanities School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Jing Luo
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Qing-Wen Tao
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|