1
|
Liu Y, Chen ZJ, Fei Y, Yu X, Chen G. Investigating the therapeutic potential of epigallocatechin gallate (EGCG) for chronic pain management: mechanisms, applications, and future perspectives. Fitoterapia 2025; 184:106646. [PMID: 40446933 DOI: 10.1016/j.fitote.2025.106646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/12/2025] [Accepted: 05/27/2025] [Indexed: 06/02/2025]
Abstract
Chronic pain, a widespread condition impacting a large segment of the global population, involves persistent and diverse pain types including nociceptive, neuropathic, and the more recently defined nociplastic pain. This review explores the potential of Epigallocatechin gallate (EGCG), a key bioactive compound in green tea, known for its extensive biological activities that aid in pain management. Highlighting its anti-inflammatory, antioxidant, and neuroprotective effects, EGCG is posited as a promising alternative to conventional pain medications, which are often associated with significant side effects and dependency risks. Despite its therapeutic promise, EGCG's clinical application is hampered by poor bioavailability and stability. By outlining the current state of research and identifying gaps for future investigation, this review underscores the importance of integrating EGCG into a broader pain management paradigm. It advocates for extensive clinical trials to confirm EGCG's efficacy and safety in humans, aiming to establish it as a part of an integrative strategy for treating chronic pain, thus potentially reducing the reliance on conventional pharmacotherapy.
Collapse
Affiliation(s)
- Yue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Zheng-Jie Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Yue Fei
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Xin Yu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China; Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China.
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China.
| |
Collapse
|
2
|
Begh MZA, Zehravi M, Reza F, Sweilam SH, Shanmugarajan TS, Arjun UVNV, Devi K, Ethiraj S, Kumar VS, Thilagam E, Fahaid Al Fahaid AA, Rab SO, Khan SL, Emran TB. Therapeutic potential of phytocompounds in rheumatoid arthritis: Molecular insights and clinical applications. Pathol Res Pract 2025; 269:155945. [PMID: 40174276 DOI: 10.1016/j.prp.2025.155945] [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: 01/14/2025] [Revised: 03/16/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by systemic involvement, inflammation, and the destruction of synovial joints. RA can be categorized as anti-citrullinated protein antibodies-positive or negative based on genetic risk factors and autoantibodies. This review systematically sourced articles related to RA, phytocompounds, signaling pathways, and clinical insights from primary medical databases, including Scopus, PubMed, and Web of Science. This review explores the therapeutic potential of phytocompounds in treating RA by targeting key inflammation and immunological response signaling pathways. Phytocompounds such as curcumin, resveratrol, and flavonoids alter essential molecular pathways in RA pathophysiology, including nuclear factor kappa-light-chain-enhancer of activated B cells, mitogen-activated protein kinases, janus kinase-signal transducer and activator of transcription, and the inflammasome. These substances possess pro-resolving, anti-apoptotic, and antioxidant properties, which enhance their therapeutic efficacy. Alternative medicine, including dietary, herbal, and nutritional supplements, may help reduce RA symptoms. In vitro, in vivo, and clinical studies have demonstrated the effectiveness of these treatments. Phytocompounds have potential as a treatment for RA by altering signaling pathways, reducing oxidative stress, and protecting cartilage and bone. However, few clinical trials confirm its long-term safety, bioavailability, and effectiveness. Further clinical trials and translational research are needed to validate the effectiveness, safety, and pharmacokinetics of phytocompounds, while identifying novel plant-derived bioactive chemicals could improve patient outcomes.
Collapse
Affiliation(s)
- Md Zamshed Alam Begh
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia.
| | - Faruk Reza
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| | - Thukani Sathanantham Shanmugarajan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai, Tamil Nadu 600117, India
| | - Uppuluri Varuna Naga Venkata Arjun
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai, Tamil Nadu 600117, India
| | - Kadirivel Devi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai, Tamil Nadu 600117, India
| | - Susithra Ethiraj
- College of Pharmacy, Sri Venkateswara University, SV University, Prakasam Rd, Sri Padmavati Mahila Visvavidyalayam, Tirupati , Andhra Pradesh, 517502, India
| | - V Santhosh Kumar
- Department of Pharmacology, Faculty of Pharmacy, ACS Medical College Campus, Dr. MGR. Educational and Research Institute, Poonamallee High Rd, Velappanchavadi, Chennai, Tamil Nadu 600077, India
| | - E Thilagam
- Department of Pharmacognosy, JKKMMRF'S-ANNAI JKK Sampooorani Ammal College of Pharmacy, Ethirmedu, Komarapalayam (Affiliated to The Tamil Nadu Dr. M.G.R. Medical University, Chennai), India
| | | | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sharuk L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa, Maharashtra 413520, India
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, USA.
| |
Collapse
|
3
|
Yue Y, Cao S, Cao F, Wei Y, Li A, Wang D, Liu P, Zeng H, Lin J. Unveiling research hotspots: a bibliometric study on macrophages in musculoskeletal diseases. Front Immunol 2025; 16:1519321. [PMID: 40356917 PMCID: PMC12066445 DOI: 10.3389/fimmu.2025.1519321] [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: 10/29/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
Research on the role of macrophages in musculoskeletal (MSK) diseases has significantly increased in recent years. However, a thorough evaluation of the developmental trajectory of this field, including the contributions of prominent authors and primary research themes, remains insufficient. Furthermore, the identification of emerging research hotspots requires more detailed exploration. This study collated articles and reviews addressing "macrophages in MSK diseases" published between 2004 and 2023, with all data extracted from the Web of Science database. The collected data were analyzed using a variety of bibliometric and visualization tools, such as VOSviewer, CiteSpace, GraphPad Prism, and R packages. Results indicate that China and the United States are the leading contributors in this research domain. Among the many academic institutions involved, Shanghai Jiao Tong University and the University of California stand out as the most productive. The journal "Frontiers in Immunology" had the highest publication output on this topic. The five most frequently explored research domains include Immunology, Rheumatology, Pharmacology and Pharmacy, Cell Biology, and Biochemistry and Molecular Biology. These results offer a comprehensive overview of the current state of research in this field and provide meaningful insights for guiding future studies.
Collapse
Affiliation(s)
- Yaohang Yue
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Shandong Second Medical University, Clinical Medical College, Weifang, China
| | - Siyang Cao
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fuyang Cao
- Department of Orthopedics, Second Hospital of Shanxi Medical University,
Taiyuan, Shanxi, China
| | - Yihao Wei
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic
University, Hong Kong, Hong Kong SAR, China
| | - Aikang Li
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Deli Wang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peng Liu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Zeng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jianjing Lin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
4
|
Xia X, He C, Xue Z, Wang Y, Qin Y, Ren Z, Huang Y, Luo H, Chen HN, Zhang WH, Huang LB, Shi Y, Bai Y, Cai B, Wang L, Zhang F, Qian M, Zhang W, Shu Y, Yin G, Xu H, Xie Q. Single cell immunoprofile of synovial fluid in rheumatoid arthritis with TNF/JAK inhibitor treatment. Nat Commun 2025; 16:2152. [PMID: 40038288 PMCID: PMC11880340 DOI: 10.1038/s41467-025-57361-0] [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: 07/13/2023] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
Numerous patients with rheumatoid arthritis (RA) manifest severe syndromes, including elevated synovial fluid volumes (SF) with abundant immune cells, which can be controlled by TNF/JAK inhibitors. Here, we apply single-cell RNA sequencing (scRNA-seq) and subsequent validations in SF from RA patients. These analyses of synovial tissue show reduced density of SF-derived pathogenic cells (e.g., SPP1+ macrophages and CXCL13+CD4+ T cells), altered gene expression (e.g., SPP1 and STAT1), molecular pathway changes (e.g., JAK/STAT), and cell-cell communications in drug-specific manners in samples from patients pre-/post-treated with adalimumab/tofacitinib. Particularly, SPP1+ macrophages exhibit pronounced communication with CXCL13+CD4+ T cells, which are abolished after treatment and correlate with treatment efficacy. These pathogenic cell types alone or in combination can augment inflammation of fibroblast-like synoviocytes in vitro, while conditional Spp1 knocking-out reduces RA-related cytokine expression in collagen-induced arthritis mice models. Our study shows the functional role of SF-derived pathogenic cells in progression and drug-specific treatment outcomes in RA.
Collapse
Affiliation(s)
- Xuyang Xia
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chenjia He
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhinan Xue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuelan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Qin
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixiang Ren
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yupeng Huang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Han Luo
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei-Han Zhang
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li-Bin Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunying Shi
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangjuan Bai
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cai
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lanlan Wang
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and Oncology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Geng Yin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Tianfu Jincheng Laboratory, Chengdu, Sichuan, China.
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
Li Z, Jiang J, Cai K, Qiao Y, Zhang X, Wang L, Kang Y, Wu X, Zhao B, Wang X, Zhang T, Lin Z, Wu J, Lu S, Gao H, Jin H, Xu C, Huangfu X, James Z, Chen Q, Zheng X, Liu NN, Zhao J. CCN2 mediates fibroblast-macrophage interaction in knee arthrofibrosis based on single-cell RNA-seq analysis. Bone Res 2025; 13:26. [PMID: 39994205 PMCID: PMC11850813 DOI: 10.1038/s41413-025-00400-9] [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: 01/08/2024] [Revised: 12/02/2024] [Accepted: 12/31/2024] [Indexed: 02/26/2025] Open
Abstract
Knee arthrofibrosis, characterized by excessive matrix protein production and deposition, substantially impairs basic daily functions, causing considerable distress and financial burden. However, the underlying pathomechanisms remain unclear. Here, we characterized the heterogeneous cell populations and cellular pathways by combination of flow cytometry and single-cell RNA-seq analysis of synovial tissues from six patients with or without knee arthrofibrosis. Increased macrophages and fibroblasts were observed with decreased numbers of fibroblast-like synoviocytes, endothelial cells, vascular smooth muscle cells, and T cells in the arthrofibrosis group compared with negative controls. Notably, fibroblasts were discovered to interact with macrophages, and lead to fibrosis through TGF-β pathway induced CCN2 expression in fibroblasts. CCN2 was demonstrated to be required for fibroblast pro-fibrotic functions (activation, proliferation, and migration) through TGFBR/SMAD pathway. The expression of CCN2 was positively correlated with the collagen volume and TGF-β expression and negatively associated with patient-reported outcome measures in another cohort of patients with knee arthrofibrosis. Our study reveals the role of CCN2 in the fibroblast-macrophage interaction through TGF-β pathway which might help to shed light on CCN2 as a potential biomarker.
Collapse
Affiliation(s)
- Ziyun Li
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jia Jiang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Kangwen Cai
- Shanghai Normal University, Shanghai, 200233, China
| | - Yi Qiao
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xuancheng Zhang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liren Wang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuhao Kang
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiulin Wu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Benpeng Zhao
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiuli Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianyi Zhang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhiqi Lin
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jinlong Wu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Simin Lu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haihan Gao
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haocheng Jin
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Caiqi Xu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaoqiao Huangfu
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhengzhi James
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qiuhua Chen
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaoqi Zheng
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jinzhong Zhao
- Department of Sports Medicine, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
6
|
Ma Y, Gong H, Cheng L, Zhang D. Discoid Domain Receptors Signaling in Macrophages-Mediated Diseases. Int J Gen Med 2025; 18:907-926. [PMID: 39990299 PMCID: PMC11847422 DOI: 10.2147/ijgm.s487093] [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: 07/15/2024] [Accepted: 01/18/2025] [Indexed: 02/25/2025] Open
Abstract
Macrophages, as a crucial component of the body's immune system, play a vital role in the onset, progression, and outcome of diseases. Discoidin domain receptors (DDRs), important members of the novel receptor tyrosine kinase superfamily, exhibit unique functions in macrophage physiology. Through interactions with the extracellular matrix, DDRs activate signaling pathways such as p38 MAPK and NF-κB, regulating macrophage adhesion, migration, and secretory functions, thereby influencing their behavior in diseases. Recent studies have indicated a direct correlation between DDRs and the progression of various diseases, including inflammation, cancer, and fibrosis. However, there remain numerous knowledge gaps regarding the specific mechanisms by which DDRs function in macrophage-mediated diseases. This article provides an in-depth summary of the regulatory mechanisms of DDRs on macrophages, detailing their modulatory roles in various diseases through macrophages and their underlying mechanisms. The aim is to offer new insights into biomedical therapies targeting DDRs and the development of novel drugs.
Collapse
Affiliation(s)
- Yaohui Ma
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Hang Gong
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Long Cheng
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| | - Dekui Zhang
- Department of Gastroenterology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, People’s Republic of China
| |
Collapse
|
7
|
Chen Z, Pang Q, Zhan J, Liu J, Zhao W, Dong L, Huang W. MSCs-derived ECM functionalized hydrogel regulates macrophage reprogramming for osteoarthritis treatment by improving mitochondrial function and energy metabolism. Mater Today Bio 2024; 29:101340. [PMID: 39640869 PMCID: PMC11617891 DOI: 10.1016/j.mtbio.2024.101340] [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: 08/10/2024] [Revised: 10/20/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease that affects the entire joint, with synovial inflammation being a major pathological feature. Macrophages, as the most abundant immune cells in the synovium, have an M1/M2 imbalance that is closely related to the occurrence and development of OA. Mesenchymal stem cells (MSCs) have been shown to effectively suppress inflammation in the treatment of OA, but they still pose issues such as immune rejection and tumorigenicity. The extracellular matrix (ECM), as a major mediator of MSCs' immunoregulatory effects, offers a cell-free therapy to circumvent these risks. In this study, we developed an ECM-functionalized hydrogel by combining MSC-derived ECM with gelatin methacryloyl (GelMA). To enhance the immunomodulatory potential of MSCs, we pre-stimulated MSCs with the inflammatory factor interleukin-6 (IL-6) present in OA. In vitro results showed that the ECM-functionalized hydrogel promoted M2 macrophage polarization and inhibited the expression of various inflammatory genes, strongly indicating the hydrogel's powerful immunoregulatory capabilities. In an in vivo rat OA model, the ECM-functionalized hydrogel significantly reduced synovial inflammation and cartilage matrix degradation, alleviating the progression of OA. Furthermore, we utilized proteomics and transcriptomics analysis to reveal that the hydrogel accomplished macrophage metabolic reprogramming by regulating mitochondrial function and energy metabolism, thereby reducing inflammation. These findings suggest that the ECM-functionalized hydrogel is a promising biomaterial-based strategy for treating OA by targeting key pathological mechanisms.
Collapse
Affiliation(s)
- Zhuolin Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Qiming Pang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jingdi Zhan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Junyan Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Weikang Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Lili Dong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Liu J, Du C, Chen H, Huang W, Lei Y. Nano-Micron Combined Hydrogel Microspheres: Novel Answer for Minimal Invasive Biomedical Applications. Macromol Rapid Commun 2024; 45:e2300670. [PMID: 38400695 DOI: 10.1002/marc.202300670] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/05/2024] [Indexed: 02/25/2024]
Abstract
Hydrogels, key in biomedical research for their hydrophilicity and versatility, have evolved with hydrogel microspheres (HMs) of micron-scale dimensions, enhancing their role in minimally invasive therapeutic delivery, tissue repair, and regeneration. The recent emergence of nanomaterials has ushered in a revolutionary transformation in the biomedical field, which demonstrates tremendous potential in targeted therapies, biological imaging, and disease diagnostics. Consequently, the integration of advanced nanotechnology promises to trigger a new revolution in the realm of hydrogels. HMs loaded with nanomaterials combine the advantages of both hydrogels and nanomaterials, which enables multifaceted functionalities such as efficient drug delivery, sustained release, targeted therapy, biological lubrication, biochemical detection, medical imaging, biosensing monitoring, and micro-robotics. Here, this review comprehensively expounds upon commonly used nanomaterials and their classifications. Then, it provides comprehensive insights into the raw materials and preparation methods of HMs. Besides, the common strategies employed to achieve nano-micron combinations are summarized, and the latest applications of these advanced nano-micron combined HMs in the biomedical field are elucidated. Finally, valuable insights into the future design and development of nano-micron combined HMs are provided.
Collapse
Affiliation(s)
- Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- 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
|
9
|
Sharma A, Sharma C, Sharma L, Wal P, Mishra P, Sachdeva N, Yadav S, Vargas De-La Cruz C, Arora S, Subramaniyan V, Rawat R, Behl T, Nandave M. Targeting the vivid facets of apolipoproteins as a cardiovascular risk factor in rheumatoid arthritis. Can J Physiol Pharmacol 2024; 102:305-317. [PMID: 38334084 DOI: 10.1139/cjpp-2023-0259] [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: 02/10/2024]
Abstract
Mostly, cardiovascular diseases are blamed for casualties in rheumatoid arthritis (RA) patients. Customarily, dyslipidemia is probably the most prevalent underlying cause of untimely demise in people suffering from RA as it hastens the expansion of atherosclerosis. The engagement of inflammatory cytokines like tumor necrosis factor-α (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), etc., is crucial in the progression and proliferation of both RA and abnormal lipid parameters. Thus, lipid abnormalities should be monitored frequently in patients with both primary and advanced RA stages. An advanced lipid profile examination, i.e., direct role of apolipoproteins associated with various lipid molecules is a more dependable approach for better understanding of the disease and selecting suitable therapeutic targets. Therefore, studying their apolipoproteins is more relevant than assessing RA patients' altered lipid profile levels. Among the various apolipoprotein classes, Apo A1 and Apo B are primarily being focused. In addition, it also addresses how calculating Apo B:Apo A1 ratio can aid in analyzing the disease's risk. The marketed therapies available to control lipid abnormalities are associated with many other risk factors. Hence, directly targeting Apo A1 and Apo B would provide a better and safer option.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Chakshu Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Lalit Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, Uttar Pradesh, India
| | - Preeti Mishra
- Raja Balwant Singh Engineering Technical Campus, Bichpuri, Agra, India
| | - Nitin Sachdeva
- Department of Anesthesia, Mediclinic Aljowhara Hospital, Al Ain, United Arab Emirates
| | - Shivam Yadav
- School of Pharmacy, Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
| | - Celia Vargas De-La Cruz
- Department of Pharmacology, Bromatology and Toxicology, Faculty of Pharmacy and Biochemistry, Universidad Nacional Mayor de San Marcos, Lima 15001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima 15001, Peru
| | - Sandeep Arora
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Ravi Rawat
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun, Uttarakhand, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Delhi, India
| |
Collapse
|
10
|
Xiao P, Han X, Huang Y, Yang J, Chen L, Cai Z, Hu N, Cui W, Huang W. Reprogramming macrophages via immune cell mobilized hydrogel microspheres for osteoarthritis treatments. Bioact Mater 2024; 32:242-259. [PMID: 37869722 PMCID: PMC10589729 DOI: 10.1016/j.bioactmat.2023.09.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Regulating macrophage activation precisely is crucial in treating chronic inflammation in osteoarthritis (OA). However, the stable pro-inflammatory state and deep distribution of macrophages in vivo pose a great challenge to treatment. In this study, inspired by the innate immune, immune cell mobilized hydrogel microspheres were constructed by microfluidic methods and load chemokines, macrophage antibodies and engineered cell membrane vesicles (sEVs) via covalent and non-covalent junctions. The immune cell mobilized hydrogel microspheres, based on a mixture of streptavidin grafted hyaluronic acid methacrylate (HAMA-SA) and Chondroitin sulfate methacrylate (ChSMA) microspheres (HCM), can recruit, capture and reprogram proinflammatory macrophages in the joint cavity to improve the joint inflammatory microenvironment. In vitro experiments demonstrated that immune cell mobilized hydrogel microspheres had excellent macrophage recruitment, capture, and reprogramming abilities. Pro-inflammatory macrophages can be transformed into anti-inflammatory macrophages with an efficiency of 88.5 %. Animal experiments also revealed significant reduction in synovial inflammation and cartilage matrix degradation of OA. Therefore, the immune cell mobilized hydrogel microspheres may be an effective treatment of OA inflammation for the future.
Collapse
Affiliation(s)
- Pengcheng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoyu Han
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Jianye Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Li Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Ning Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Orthopaedic Research Laboratory, Chongqing Medical University, 400016, Chongqing, China
| |
Collapse
|
11
|
Hu H, Dou X, Hu X, Wang L, Ma Y, Liu J, Zhou X, Cao H, Liu X, Deng X, Li N. Identification of a novel cuproptosis-related gene signature for rheumatoid arthritis-A prospective study. J Gene Med 2023; 25:e3535. [PMID: 37338187 DOI: 10.1002/jgm.3535] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a multifactorial systemic autoimmune disease characterized by ongoing synovial inflammation, leading to the degradation of cartilage. Cuproptosis, as a newly characterized form of cell death, may influence RA progression by regulating immune cells and chondrocytes. This study sets out to identify the hub cuproptosis-related gene (CRG) associated with the pathogenesis of RA. METHODS A series of bioinformatic analyses were performed to evaluate the expression score of CRGs and the immune infiltration landscape between RA and normal samples. The hub gene was screened through the correlation analysis of CRGs, and the interaction network between the hub gene and transcription factors (TFs) was constructed. Finally, the hub gene was validated through quantitative real-time polymerase chain reaction (qRT-PCR) of patient samples and cell experiments. RESULTS Drolipoamide S-acetyltransferase (DLAT) was screened as the hub gene. Correlation analysis between the hub gene and immune microenvironment demonstrated that DLAT had the highest correlation with T follicular helper cells. Eight pairs of DLAT-TF interaction networks were constructed. Single-cell sequencing showed that CRGs were highly expressed in RA chondrocytes, and chondrocytes could be classified into three different subsets. qRT-PCR was used to validate the above results. Dlat knockdown in immortalized human chondrocytes led to significantly improved mitochondrial membrane potentials and reduced levels of intracellular reactive oxygen species (ROS), mitochondrial ROS and apoptosis. CONCLUSIONS This study rudimentarily demonstrates the correlation between CRGs and immune cell infiltration in RA. The biomarker DLAT may provide comprehensive insights into the pathogenesis and drug targets of RA.
Collapse
Affiliation(s)
- Han Hu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xinyu Dou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Xiangjia Hu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Linbang Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Yunlong Ma
- Pain Medicine Center, Peking University Third Hospital, Beijing, China
| | - Jingkun Liu
- Data Centre Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xuchang Zhou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Hong Cao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xiaoguang Liu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Nan Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| |
Collapse
|
12
|
Wang P, Zhang Y, Lei H, Yu J, Zhou Q, Shi X, Zhu Y, Zhang D, Zhang P, Wang K, Dong K, Xing J, Dong Y. Hyaluronic acid-based M1 macrophage targeting and environmental responsive drug releasing nanoparticle for enhanced treatment of rheumatoid arthritis. Carbohydr Polym 2023; 316:121018. [PMID: 37321721 DOI: 10.1016/j.carbpol.2023.121018] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023]
Abstract
Herein, hyaluronic acid (HA) and β-cyclodextrin (β-CD) is used to form targeted drug delivery platform HCPC/DEX NPs with previously prepared carbon dots (CDs) as cross-linker, dexamethasone (DEX) is loaded for rheumatoid arthritis (RA) treatment. The drug loading capacity of β-CD and M1 macrophage targeting of HA were utilized for efficient delivery of DEX to the inflammatory joints. Because of the environmental responsive degradation of HA, DEX can be released in 24 h and inhibit the inflammatory response in M1 macrophages. The drug loading of NPs is 4.79 %. Cellular uptake evaluation confirmed that NPs can specifically target to M1 macrophages via HA ligands, the uptake of M1 macrophages is 3.7 times that of normal macrophages. In vivo experiments revealed that NPs can accumulate in RA joints to alleviate inflammation and accelerate cartilage healing, the accumulation can be observed in 24 h. The cartilage thickness increased to 0.45 mm after HCPC/DEX NPs treatment, indicating its good RA therapeutic effect. Importantly, this study was the first to utilize the potential acid and reactive oxygen species responsiveness of HA to release drug and prepare M1 macrophage targeting nanodrug for RA treatment, which provides a safe and effective RA therapeutic strategy.
Collapse
Affiliation(s)
- Pengchong Wang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hengyu Lei
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jie Yu
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qinyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xianpeng Shi
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Yaning Zhu
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Dan Zhang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Peng Zhang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Ke Wang
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kai Dong
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Jianfeng Xing
- Department of Pharmaceutics, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yalin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
13
|
Philippon EML, van Rooijen LJE, Khodadust F, van Hamburg JP, van der Laken CJ, Tas SW. A novel 3D spheroid model of rheumatoid arthritis synovial tissue incorporating fibroblasts, endothelial cells, and macrophages. Front Immunol 2023; 14:1188835. [PMID: 37545512 PMCID: PMC10402919 DOI: 10.3389/fimmu.2023.1188835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Objective Rheumatoid Arthritis (RA) is a progressive and systemic autoimmune disorder associated with chronic and destructive joint inflammation. The hallmarks of joint synovial inflammation are cellular proliferation, extensive neoangiogenesis and infiltration of immune cells, including macrophages. In vitro approaches simulating RA synovial tissue are crucial in preclinical and translational research to evaluate novel diagnostic and/or therapeutic markers. Two-dimensional (2D) settings present very limited in vivo physiological proximity as they cannot recapitulate cell-cell and cell-matrix interactions occurring in the three-dimensional (3D) tissue compartment. Here, we present the engineering of a spheroid-based model of RA synovial tissue which mimics 3D interactions between cells and pro-inflammatory mediators present in the inflamed synovium. Methods Spheroids were generated by culturing RA fibroblast-like-synoviocytes (RAFLS), human umbilical vein endothelial cells (ECs) and monocyte-derived macrophages in a collagen-based 3D scaffold. The spheroids were cultured in the presence or absence of vascular endothelial growth factor (VEGF) and fibroblast growth factor 2 (bFGF) or RA synovial fluid (SF). Spheroid expansion and cell migration were quantified for all conditions using confocal microscopy and digital image analysis. Results A novel approach using machine learning was developed to quantify spheroid outgrowth and used to reexamine the existing spheroid-based model of RA synovial angiogenesis consisting of ECs and RAFLS. A 2-fold increase in the spheroid outgrowth ratio was demonstrated upon VEGF/bFGF stimulation (p<0.05). The addition of macrophages within the spheroid structure (3.75x104 RAFLS, 7.5x104 ECs and 3.0x104 macrophages) resulted in good incorporation of the new cell type. The addition of VEGF/bFGF significantly induced spheroid outgrowth (p<0.05) in the new system. SF stimulation enhanced containment of macrophages within the spheroids. Conclusion We present a novel spheroid based model consisting of RAFLS, ECs and macrophages that reflects the RA synovial tissue microenvironment. This model may be used to dissect the role of specific cell types in inflammatory responses in RA, to study specific signaling pathways involved in the disease pathogenesis and examine the effects of novel diagnostic (molecular imaging) and therapeutic compounds, including small molecule inhibitors and biologics.
Collapse
Affiliation(s)
- Eva M. L. Philippon
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Lisanne J. E. van Rooijen
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Fatemeh Khodadust
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Conny J. van der Laken
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
Tian L, Zhang Z, Mao Y, Zong M. Association between pregnant women with rheumatoid arthritis and preeclampsia: A systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e34131. [PMID: 37390281 PMCID: PMC10313298 DOI: 10.1097/md.0000000000034131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND To examine the association between pregnant women with rheumatoid arthritis (RA) and the risk of preeclampsia. METHODS This study was registered on the International Prospective Register of Systematic Reviews (PROSPERO) under the number CRD42022361571. The primary outcome was preeclampsia. Two evaluators independently reviewed the included studies, assessed their risk of bias, and extracted the data. Unadjusted and adjusted ratios with 95% confidence intervals and 95% prediction intervals were calculated. Heterogeneity was quantified using the І2 statistic, where І2 ≥ 50% indicated the presence of significant heterogeneity. Subgroup and sensitivity analyses were performed to test the robustness of the overall findings. RESULTS A total of 8 studies, including 10,951,184 pregnant women, of whom 13,333 were diagnosed with RA, met the inclusion criteria. Meta-analysis revealed that pregnant women with RA were significantly more likely to develop preeclampsia than those without RA (pooled odds ratio, 1.66; 95% confidence interval, 1.52-1.80; P < .001; І2 < .001). CONCLUSION RA during pregnancy is associated with higher odds of preeclampsia.
Collapse
Affiliation(s)
- Lv Tian
- School of Nursing, Jilin University, Changchun, China
| | - Zhiyuan Zhang
- School of Nursing, Jilin University, Changchun, China
| | - Yuting Mao
- Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| | - Minru Zong
- School of Nursing, Jilin University, Changchun, China
- Department of Rehabilitation, The Third Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Radu AF, Bungau SG. Nanomedical approaches in the realm of rheumatoid arthritis. Ageing Res Rev 2023; 87:101927. [PMID: 37031724 DOI: 10.1016/j.arr.2023.101927] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Rheumatoid arthritis (RA) is a heterogeneous autoimmune inflammatory disorder defined by the damage to the bone and cartilage in the synovium, which causes joint impairment and an increase in the mortality rate. It is associated with an incompletely elucidated pathophysiological mechanism. Even though disease-modifying antirheumatic drugs have contributed to recent improvements in the standard of care for RA, only a small fraction of patients is able to attain and maintain clinical remission without the necessity for ongoing immunosuppressive drugs. The evolution of tolerance over time as well as patients' inability to respond to currently available therapy can alter the overall management of RA. A significant increase in the research of RA nano therapies due to the possible improvements they may provide over traditional systemic treatments has been observed. New approaches to getting beyond the drawbacks of existing treatments are presented by advancements in the research of nanotherapeutic techniques, particularly drug delivery nano systems. Via passive or active targeting of systemic delivery, therapeutic drugs can be precisely transported to and concentrated in the affected sites. As a result, nanoscale drug delivery systems improve the solubility and bioavailability of certain drugs and reduce dose escalation. In the present paper, we provide a thorough overview of the possible biomedical applications of various nanostructures in the diagnostic and therapeutic management of RA, derived from the shortcomings of conventional therapies. Moreover, the paper suggests the need for improvement on the basis of research directions and properly designed clinical studies.
Collapse
Affiliation(s)
- Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| |
Collapse
|
16
|
Liu Q, Wang X, Chen Y, Ma X, Kang X, He F, Feng D, Zhang Y. Ablation of myeloid discoidin domain receptor 2 exacerbates arthritis and high fat diet induced inflammation. Biochem Biophys Res Commun 2023; 649:47-54. [PMID: 36745969 DOI: 10.1016/j.bbrc.2023.01.074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/22/2023] [Indexed: 02/04/2023]
Abstract
Chronic systemic inflammation leads to sever disorders and diseases. It is of great importance to explore novel target for effective treatment. Discoidin domain receptor 2 (Ddr2) is a member of receptor tyrosine kinase (RTK) family and is implicated in skeletal and fat hemostasis. However, the role of Ddr2 in myeloid cells remains obscure. In this study, we conditionally deleted Ddr2 in myeloid lineage cells to generate cKO mice to investigate the role of Ddr2 in myeloid lineage cells. We found that cKO mice exhibited more severe inflammation both in collagen antibody-induced arthritis (CAIA) and high-fat diet (HFD)-induced obesity, indicating the protective role of Ddr2 against inflammation. Mechanistically, Ddr2 promotes macrophage repolarization from the M1 to M2 phenotype, and protect against systemic inflammation. Our study reveals for the first time that Ddr2 modulates macrophage repolarization and plays critical roles in macrophage-mediated inflammation, providing potential target for the intervention of inflammation and related diseases.
Collapse
Affiliation(s)
- Qingyun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaolong Wang
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yazhuo Chen
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiao Ma
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaomin Kang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Fang He
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Dongxu Feng
- Department of Orthopaedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, People's Republic of China.
| | - Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
17
|
Semenistaja S, Skuja S, Kadisa A, Groma V. Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk. Int J Mol Sci 2023; 24:4120. [PMID: 36835530 PMCID: PMC9964755 DOI: 10.3390/ijms24044120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive, severely debilitating, and multifactorial joint disease that is recognized as the most common type of arthritis. During the last decade, it shows an incremental global rise in prevalence and incidence. The interaction between etiologic factors that mediate joint degradation has been explored in numerous studies. However, the underlying processes that induce OA remain obscure, largely due to the variety and complexity of these mechanisms. During synovial joint dysfunction, the osteochondral unit undergoes cellular phenotypic and functional alterations. At the cellular level, the synovial membrane is influenced by cartilage and subchondral bone cleavage fragments and extracellular matrix (ECM) degradation products from apoptotic and necrotic cells. These "foreign bodies" serve as danger-associated molecular patterns (DAMPs) that trigger innate immunity, eliciting and sustaining low-grade inflammation in the synovium. In this review, we explore the cellular and molecular communication networks established between the major joint compartments-the synovial membrane, cartilage, and subchondral bone of normal and OA-affected joints.
Collapse
Affiliation(s)
- Sofija Semenistaja
- Department of Doctoral Studies, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Sandra Skuja
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Anda Kadisa
- Department of Internal Diseases, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Valerija Groma
- Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
| |
Collapse
|
18
|
Organokines in Rheumatoid Arthritis: A Critical Review. Int J Mol Sci 2022; 23:ijms23116193. [PMID: 35682868 PMCID: PMC9180954 DOI: 10.3390/ijms23116193] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease that primarily affects the joints. Organokines can produce beneficial or harmful effects in this condition. Among RA patients, organokines have been associated with increased inflammation and cartilage degradation due to augmented cytokines and metalloproteinases production, respectively. This study aimed to perform a review to investigate the role of adipokines, osteokines, myokines, and hepatokines on RA progression. PubMed, Embase, Google Scholar, and Cochrane were searched, and 18 studies were selected, comprising more than 17,000 RA patients. Changes in the pattern of organokines secretion were identified, and these could directly or indirectly contribute to aggravating RA, promoting articular alterations, and predicting the disease activity. In addition, organokines have been implicated in higher radiographic damage, immune dysregulation, and angiogenesis. These can also act as RA potent regulators of cells proliferation, differentiation, and apoptosis, controlling osteoclasts, chondrocytes, and fibroblasts as well as immune cells chemotaxis to RA sites. Although much is already known, much more is still unknown, principally about the roles of organokines in the occurrence of RA extra-articular manifestations.
Collapse
|
19
|
Ajalik RE, Alenchery RG, Cognetti JS, Zhang VZ, McGrath JL, Miller BL, Awad HA. Human Organ-on-a-Chip Microphysiological Systems to Model Musculoskeletal Pathologies and Accelerate Therapeutic Discovery. Front Bioeng Biotechnol 2022; 10:846230. [PMID: 35360391 PMCID: PMC8964284 DOI: 10.3389/fbioe.2022.846230] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Human Microphysiological Systems (hMPS), otherwise known as organ- and tissue-on-a-chip models, are an emerging technology with the potential to replace in vivo animal studies with in vitro models that emulate human physiology at basic levels. hMPS platforms are designed to overcome limitations of two-dimensional (2D) cell culture systems by mimicking 3D tissue organization and microenvironmental cues that are physiologically and clinically relevant. Unlike animal studies, hMPS models can be configured for high content or high throughput screening in preclinical drug development. Applications in modeling acute and chronic injuries in the musculoskeletal system are slowly developing. However, the complexity and load bearing nature of musculoskeletal tissues and joints present unique challenges related to our limited understanding of disease mechanisms and the lack of consensus biomarkers to guide biological therapy development. With emphasis on examples of modeling musculoskeletal tissues, joints on chips, and organoids, this review highlights current trends of microphysiological systems technology. The review surveys state-of-the-art design and fabrication considerations inspired by lessons from bioreactors and biological variables emphasizing the role of induced pluripotent stem cells and genetic engineering in creating isogenic, patient-specific multicellular hMPS. The major challenges in modeling musculoskeletal tissues using hMPS chips are identified, including incorporating biological barriers, simulating joint compartments and heterogenous tissue interfaces, simulating immune interactions and inflammatory factors, simulating effects of in vivo loading, recording nociceptors responses as surrogates for pain outcomes, modeling the dynamic injury and healing responses by monitoring secreted proteins in real time, and creating arrayed formats for robotic high throughput screens. Overcoming these barriers will revolutionize musculoskeletal research by enabling physiologically relevant, predictive models of human tissues and joint diseases to accelerate and de-risk therapeutic discovery and translation to the clinic.
Collapse
Affiliation(s)
- Raquel E. Ajalik
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Rahul G. Alenchery
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - John S. Cognetti
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Benjamin L. Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Department of Dermatology, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- *Correspondence: Hani A. Awad,
| |
Collapse
|
20
|
Haubruck P, Pinto MM, Moradi B, Little CB, Gentek R. Monocytes, Macrophages, and Their Potential Niches in Synovial Joints - Therapeutic Targets in Post-Traumatic Osteoarthritis? Front Immunol 2021; 12:763702. [PMID: 34804052 PMCID: PMC8600114 DOI: 10.3389/fimmu.2021.763702] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Synovial joints are complex structures that enable normal locomotion. Following injury, they undergo a series of changes, including a prevalent inflammatory response. This increases the risk for development of osteoarthritis (OA), the most common joint disorder. In healthy joints, macrophages are the predominant immune cells. They regulate bone turnover, constantly scavenge debris from the joint cavity and, together with synovial fibroblasts, form a protective barrier. Macrophages thus work in concert with the non-hematopoietic stroma. In turn, the stroma provides a scaffold as well as molecular signals for macrophage survival and functional imprinting: “a macrophage niche”. These intricate cellular interactions are susceptible to perturbations like those induced by joint injury. With this review, we explore how the concepts of local tissue niches apply to synovial joints. We introduce the joint micro-anatomy and cellular players, and discuss their potential interactions in healthy joints, with an emphasis on molecular cues underlying their crosstalk and relevance to joint functionality. We then consider how these interactions are perturbed by joint injury and how they may contribute to OA pathogenesis. We conclude by discussing how understanding these changes might help identify novel therapeutic avenues with the potential of restoring joint function and reducing post-traumatic OA risk.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Marlene Magalhaes Pinto
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Babak Moradi
- Clinic of Orthopaedics and Trauma Surgery, University Clinic of Schleswig-Holstein, Kiel, Germany
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Radu AF, Bungau SG. Management of Rheumatoid Arthritis: An Overview. Cells 2021; 10:2857. [PMID: 34831081 PMCID: PMC8616326 DOI: 10.3390/cells10112857] [Citation(s) in RCA: 449] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease of unknown etiology, primarily affecting the joints, then extra-articular manifestations can occur. Due to its complexity, which is based on an incompletely elucidated pathophysiological mechanism, good RA management requires a multidisciplinary approach. The clinical status of RA patients has improved in recent years due to medical advances in diagnosis and treatment, that have made it possible to reduce disease activity and prevent systemic complications. The most promising results were obtained by developing disease-modifying anti-rheumatic drugs (DMARDs), the class to which conventional synthetic, biologic, and targeted synthetic drugs belong. Furthermore, ongoing drug development has led to obtaining molecules with improved efficacy and safety profiles, but further research is needed until RA turns into a curable pathology. In the present work, we offer a comprehensive perspective on the management of RA, by centralizing the existing data provided by significant literature, emphasizing the importance of an early and accurate diagnosis associated with optimal personalized treatment in order to achieve better outcomes for RA patients. In addition, this study suggests future research perspectives in the treatment of RA that could lead to higher efficacy and safety profiles and lower financial costs.
Collapse
Affiliation(s)
- Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|