1
|
Zhang SP, Tong M, Mo J, Dong ZY, Huang YF. M2 macrophages activate the IL-10/JAK2/STAT3 pathway to induce pathological microangiogenesis in the nucleus pulposus exacerbating intervertebral disc degeneration. J Orthop Surg Res 2025; 20:532. [PMID: 40426248 DOI: 10.1186/s13018-025-05962-2] [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: 04/05/2025] [Accepted: 05/23/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Macrophage infiltration accompanied by pathological microangiogenesis in the nucleus pulposus (NP) plays a critical role in the progression of intervertebral disc degeneration (IDD). However, the involvement of M2 macrophages in mediating NP pathological angiogenesis and their underlying mechanisms remain unclear. METHODS Firstly, the expression of M2 macrophage (CD206) and microangiogenic (CD34) markers in human degenerated NP was observed by immunohistochemical staining, subsequently, a co-culture system of M2 macrophages and NP cells was established. IL-10 expression was silenced using siRNA to assess the pro-angiogenic effects of M2 macrophages in IDD via IL-10 and its downstream janus kinase (JAK) 2/ signal transducer and activator of transcription (STAT) 3 pathway. AG490, a specific JAK2/STAT3 inhibitor, was applied to determine whether IL-10 exerts its effects through this pathway and to evaluate its impact on angiogenesis and extracellular matrix (ECM) metabolism in NP pathology. RESULTS CD206 and CD34 were co-expressed in degenerated NP tissue. Degenerated NP cells secreted CCL17, CCL18, and CD206, exhibiting M2-like characteristics. Co-culture of M2 macrophages with degenerated NP cells led to IL-10 secretion to promote CD34 expression, and downregulated anabolic genes (type II collagen (COL2), aggrecan), and upregulated catabolic genes (matrix metalloproteinase (MMP)-3, MMP-7). JAK2 and STAT3 expression was significantly increased following co-culture. Activation of the JAK2/STAT3 pathway enhanced vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor (VEGFR), and CD34 expression and induced further downregulation of COL2 and aggrecan and upregulation of MMP-3 and MMP-7. CONCLUSION M2 macrophage infiltration and pathological neovascularization are prominent in degenerated NP tissue. IL-10 secreted by M2 macrophages activates the JAK2/STAT3 pathway to promote pathological microangiogenesis by up-regulate the expression of VEGF/VEGFR. This process disrupts ECM and accelerates the progression of IDD. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Si-Ping Zhang
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Min Tong
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Jun Mo
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Zhen-Yu Dong
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China
| | - Yi-Fei Huang
- The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China.
- Department of Spinal Surgery, Traditional Chinese Medicine Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830000, P.R. China.
- Xinjiang Uygur Autonomous Region Academy of Traditional Chinese Medicine, Urumqi, Xinjiang, 830000, P.R. China.
| |
Collapse
|
2
|
Li H, Liu H, Zhou Y, Cheng L, Wang B, Ma J. The multifaceted roles of extracellular vesicles in osteonecrosis of the femoral head. J Orthop Translat 2025; 52:70-84. [PMID: 40256260 PMCID: PMC12008682 DOI: 10.1016/j.jot.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a severe disease characterized by bone tissue necrosis due to vascular impairment, often leading to joint collapse and requiring surgical intervention. Extracellular vesicles (EVs) serve as crucial mediators of intercellular communication, influencing osteogenesis, angiogenesis, and immune regulation. This review summarizes the dual role of EVs in both the pathogenesis of ONFH and post-necrosis bone repair, highlighting the impact of various EV-mediated signaling pathways on bone regeneration and the potential crosstalk among these pathways. Additionally, EVs hold promise as diagnostic biomarkers or contrast agents to complement conventional imaging techniques for ONFH detection. By elucidating the role of EVs in osteonecrosis and addressing the current challenges, we aspire to establish a foundation for the timely identification and treatment of ONFH. The translational potential of this article: This review comprehensively discusses the role of EVs in ONFH, providing innovative and promising insights for its diagnosis and treatment, which also establishes a theoretical foundation for the future clinical application of EVs in ONFH.
Collapse
Affiliation(s)
- Hongxu Li
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Haoyang Liu
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Yu Zhou
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Liming Cheng
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Bailiang Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jinhui Ma
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
3
|
Wu L, Cao X, Shen B. Development of a macrophage polarization-modulating therapeutic agent for osteoarthritis treatment. J Orthop Surg Res 2025; 20:279. [PMID: 40082923 PMCID: PMC11908040 DOI: 10.1186/s13018-025-05679-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative joint disease. Recent studies have emphasized the crucial role of macrophages, particularly tissue-resident macrophages (Tissue-Resident Macrophages, TRMs), in the pathogenesis and progression of OA. Under physiological conditions, TRMs maintain joint homeostasis, but under various stimuli, they can polarize into pro-inflammatory M1 or anti-inflammatory M2 phenotypes. An imbalance in macrophage polarization, favoring the M1 phenotype, leads to sustained inflammation, cartilage degradation, and osteophyte formation, further exacerbating OA symptoms and structural damage. This article reviews the current understanding of macrophage polarization in OA, with a particular emphasis on the mechanisms by which TRMs influence the joint microenvironment. It explores the therapeutic potential of drug molecular platforms aimed at regulating macrophage polarization, shifting the balance from pro-inflammatory M1 to anti-inflammatory M2. The discussion includes various pharmacological agents such as corticosteroids, hyaluronic acid derivatives, monoclonal antibodies, and bioactive molecules like Squid Type II Collagen (SCII) in modulating macrophage function and slowing OA progression. Additionally, the article examines advancements in gene therapy methods targeting macrophages, utilizing nanotechnology-based delivery systems to enhance the specificity and efficiency of macrophage phenotype regulation. Targeting TRMs through sophisticated drug molecular platforms presents a promising strategy for developing novel diagnostic and therapeutic interventions for osteoarthritis.
Collapse
Affiliation(s)
- Limin Wu
- Department of Orthopaedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaotao Cao
- Early-Phase Clinical Research Unit, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Shen
- Department of Orthopaedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Zhang Y, Yan W, Wu L, Yu Z, Quan Y, Xie X. Different exosomes are loaded in hydrogels for the application in the field of tissue repair. Front Bioeng Biotechnol 2025; 13:1545636. [PMID: 40099037 PMCID: PMC11911322 DOI: 10.3389/fbioe.2025.1545636] [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: 12/15/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Exosomes are double-membrane vesicular nanoparticles in the category of extracellular vesicles, ranging in size from 30 to 150 nm, and are released from cells through a specific multi-step exocytosis process. Exosomes have emerged as promising tools for tissue repair due to their ability to transfer bioactive molecules that promote cell proliferation, differentiation, and tissue regeneration. However, the therapeutic application of exosomes is hindered by their rapid clearance from the body and limited retention at the injury site. To overcome these challenges, hydrogels, known for their high biocompatibility and porous structure, have been explored as carriers for exosomes. Hydrogels can provide a controlled release mechanism, prolonging the retention time of exosomes at targeted tissues, thus enhancing their therapeutic efficacy. This review focuses on the combination of different exosomes with hydrogels in the context of tissue repair. We first introduce the sources and functions of exosomes, particularly those from mesenchymal stem cells, and their roles in regenerative medicine. We then examine various types of hydrogels, highlighting their ability to load and release exosomes. Several strategies for encapsulating exosomes in hydrogels are discussed, including the impact of hydrogel composition and structure on exosome delivery efficiency. Finally, we review the applications of exosomes-loaded hydrogels in the repair of different tissues, such as skin, bone, cartilage, and nerve, and explore the challenges and future directions in this field. The combination of exosomes with hydrogels offers significant promise for advancing tissue repair strategies and regenerative therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Xie
- College of Life Sciences, Northwest University, Xi’an, China
| |
Collapse
|
5
|
Zhu K, Liu W, Peng Y, Wang X, Wang Z, Zheng J, Deng G, Wang Q. Study on the mechanism of Shuanghe decoction against steroid-induced osteonecrosis of the femoral head: insights from network pharmacology, metabolomics, and gut microbiota. J Orthop Surg Res 2025; 20:202. [PMID: 40001178 PMCID: PMC11863617 DOI: 10.1186/s13018-025-05619-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Steroid-induced osteonecrosis of the femoral head (SONFH) is a challenging and debilitating orthopedic condition with a rising incidence in recent years. Shuanghe Decoction (SHD), a traditional Chinese medicine formula, has shown significant efficacy in treating SONFH, though its underlying mechanisms remain unclear. PURPOSE This study aims to elucidate the therapeutic effects and potential mechanisms of SHD on SONFH through in vivo experiments, combined with network pharmacology, metabolomics, and gut microbiota analysis. MATERIALS AND METHODS Forty male Sprague-Dawley rats (300 ± 20 g) were randomly assigned to four groups: Control, Model, SHD-L, and SHD-H, with 10 rats each. SONFH was induced in all groups except the Control group using lipopolysaccharide and methylprednisolone. The SHD-L and SHD-H groups were treated with Shuanghe decoction at doses of 4.86 g/kg/day and 9.72 g/kg/day, respectively, for eight weeks. Bone morphology, pathological changes, and osteogenic factors were evaluated using Micro-CT, histological staining, and immunohistochemistry. Network pharmacology, metabolomics, and gut microbiota analyses were conducted to explore SHD's mechanisms. RESULTS SHD improved bone morphology and increased osteogenic factor expression (RUNX2, OCN, COL-I). Network pharmacology indicated that metabolic pathways play a key role in SHD's therapeutic effects. Metabolomic analysis identified 14 differential metabolites, including 21-hydroxypregnenolone and tyramine, which were restored to normal levels by SHD. Gut microbiota analysis revealed that SHD modulated bacterial abundance, particularly Verrucomicrobia, Allobaculum, and Burkholderiales. A comprehensive network identified two key metabolites (tyramine, 21-hydroxypregnenolone), seven targets (CYP19A1, CYP1A2, CYP1B1, CYP2C9, CYP3A4, MIF, and HSD11B1), two metabolic pathways (tyrosine metabolism, steroid hormone biosynthesis), and four bacterial taxa (Jeotgalicoccus, Clostridium, Corynebacterium, rc4-4) as central to SHD against SONFH. CONCLUSION SHD alleviates SONFH by reshaping gut microbiota, reversing metabolic imbalances, and enhancing osteogenesis. Our findings provide novel insights into the pharmacological mechanisms of SHD, laying a foundation for its clinical application in treating SONFH.
Collapse
Affiliation(s)
- Kai Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China
| | - Wanxin Liu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China
| | - Yuanyuan Peng
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China
| | - Xiaoqiang Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China
| | - Zhenhao Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China
| | - Jun Zheng
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China.
| | - Guoying Deng
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, P.R. China.
| | - Qiugen Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, P.R. China.
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, P.R. China.
| |
Collapse
|
6
|
Fan J, Xie Y, Liu D, Cui R, Zhang W, Shen M, Cao L. Crosstalk Between H-Type Vascular Endothelial Cells and Macrophages: A Potential Regulator of Bone Homeostasis. J Inflamm Res 2025; 18:2743-2765. [PMID: 40026304 PMCID: PMC11871946 DOI: 10.2147/jir.s502604] [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: 10/23/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
The crosstalk between H-type endothelial cells (ECs) and macrophages is critical for maintaining angiogenesis and osteogenesis in bone homeostasis. As core components of type H vessels, ECs respond to various pro-angiogenic signals, forming specialized vascular structures characterized by high expression of platelet-endothelial cell adhesion molecule-1 (CD31) and endothelial mucin (EMCN), thereby facilitating angiogenesis-osteogenesis coupling during bone formation. Macrophages, as key immune cells in the perivascular region, are primarily classified into the classically activated pro-inflammatory M1 phenotype and the selectively activated anti-inflammatory M2 phenotype, thereby performing dual functions in regulating local tissue homeostasis and innate immunity. In recent years, the complex crosstalk between type H vessel ECs and macrophages has garnered significant interest in the context of bone-related diseases. Orderly regulation of angiogenesis and bone immunity provides a new direction for preventing bone metabolic disorders such as osteoporosis and osteoarthritis. However, their interactions in bone homeostasis remain insufficiently understood, with limited clinical data available. This review comprehensively examines the intricate interactions between type H vessel ECs and macrophages with diverse phenotypes, and Insights into the signaling pathways that regulate their crosstalk, focusing on their roles in angiogenesis and osteogenesis. Furthermore, the review discusses recent interventions targeting this crosstalk and the challenges that remain. These insights may offer new perspectives on bone homeostasis and provide a theoretical foundation for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiaxuan Fan
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yaohui Xie
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Desun Liu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Rui Cui
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Wei Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Mengying Shen
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Linzhong Cao
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| |
Collapse
|
7
|
Sun Y, Zhang W, Luo Z, Zhu C, Zhang Y, Shu Z, Shen C, Yao X, Wang Y, Wang X. ZnO‐CuS/F127 Hydrogels with Multienzyme Properties for Implant‐Related Infection Therapy by Inhibiting Bacterial Arginine Biosynthesis and Promoting Tissue Repair. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202415778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Indexed: 02/08/2025]
Abstract
AbstractImplant‐related infections are characterized by the formation of bacterial biofilms. Current treatments have various drawbacks. Nanozymes with enzyme‐like activity can produce highly toxic substances to kill bacteria and remove biofilms without inducing drug resistance. However, it is difficult for current monometallic nanozymes to function well in complex biofilm environments. Therefore, the development of multimetallic nanozymes with efficient multienzyme activities is crucial. In the present study, bimetallic nanozyme, ZnO‐CuS nanoflowers with peroxidase (POD), glutathione oxidase (GSH‐Px), and catalase (CAT) activity are successfully synthesized via calcination and loaded into F127 hydrogels for the treatment of implant‐related infections. The ability of ZnO‐CuS nanoflowers to bind bacteria is key for efficient antimicrobial activity. In addition, ZnO‐CuS nanoflowers with H2O2 disrupt the metabolism of MRSA, including arginine synthesis, nucleotide excision repair, energy metabolism, and protein synthesis. ZnO‐CuS/F127 hydrogel in combination with H2O2 has been demonstrated to be effective in clearing biofilm infection and facilitating the switch of M1 macrophages to M2‐repairative phenotype macrophages for the treatment of implant infections in mice. Furthermore, ZnO‐CuS/F127 hydrogels have favorable biosafety, and their toxicity is negligible. ZnO‐CuS/F127 hydrogel has provided a promising biomedical strategy for the healing of implant‐related infections, highlighting the potential of bimetallic nanozymes for clinical applications.
Collapse
Affiliation(s)
- Yiwei Sun
- College and Hospital of Stomatology Key Lab. of Oral Diseases Research of Anhui Province Anhui Medical University Hefei 230032 P. R. China
- Department of Orthopedics The First Affiliated Hospital of Anhui Medical University Anhui Medical University Hefei 230022 P. R. China
| | - Wei Zhang
- School of Biomedical Engineering Research and Engineering Center of Biomedical Materials Anhui Medical University Hefei 230032 China
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital Fudan University Shanghai 200040 P. R. China
| | - Can Zhu
- School of Biomedical Engineering Research and Engineering Center of Biomedical Materials Anhui Medical University Hefei 230032 China
| | - Yiqun Zhang
- Department of Orthopedics The First Affiliated Hospital of Anhui Medical University Anhui Medical University Hefei 230022 P. R. China
| | - Zheng Shu
- Joint Key Laboratory of the Ministry of Education Institute of Applied Physics and Materials Engineering University of Macau Taipa Macau 999078 China
| | - Cailiang Shen
- Department of Orthopedics The First Affiliated Hospital of Anhui Medical University Anhui Medical University Hefei 230022 P. R. China
| | - Xiaxi Yao
- School of Chemistry and Materials Engineering Suzhou Key Laboratory of Functional Ceramic Materials Changshu Institute of Technology Changshu 215500 P. R. China
| | - Yuanyin Wang
- College and Hospital of Stomatology Key Lab. of Oral Diseases Research of Anhui Province Anhui Medical University Hefei 230032 P. R. China
- Department of Orthopedics The First Affiliated Hospital of Anhui Medical University Anhui Medical University Hefei 230022 P. R. China
| | - Xianwen Wang
- College and Hospital of Stomatology Key Lab. of Oral Diseases Research of Anhui Province Anhui Medical University Hefei 230032 P. R. China
- School of Biomedical Engineering Research and Engineering Center of Biomedical Materials Anhui Medical University Hefei 230032 China
| |
Collapse
|
8
|
Cai Z, Shu L, Wang C, Xie X, Liu X. M2 Macrophage-Derived Exosomes Promote Tendon-to-Bone Healing by Alleviating Cellular Senescence in Aged Rats. Arthroscopy 2024:S0749-8063(24)00737-0. [PMID: 39326562 DOI: 10.1016/j.arthro.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024]
Abstract
PURPOSE To explore the potential of M2 macrophage-derived exosomes (M2-Exos) in enhancing tendon-to-bone healing in aged rats by mitigating cellular senescence of bone marrow-derived stem cells (BMSCs). METHODS In vitro, the effects of M2-Exos on alleviating cellular senescence and improving chondrogenic potential of senescent BMSCs were evaluated. Rats (24 young and 48 aged) with chronic rotator cuff tear (RCT) were repaired and assigned into 3 groups: young group (young rats injected with fibrin at the enthesis), aged group (aged rats injected with fibrin at the enthesis), and aged + M2-Exos group (aged rats injected with fibrin containing M2-Exos at the enthesis). At 6 and 12 weeks after repair, enthesis regeneration was evaluated. Proteomic analysis was conducted to explore the mechanism through which M2-Exos mitigated cellular senescence. RESULTS In senescent BMSCs treated with M2-Exos, there was a reduction in senescence biomarkers including senescence-associated β-galactosidase, p53, p21, and senescence-associated secretory phenotype (P < .001). M2-Exos also enhanced chondrogenic potential of senescent BMSCs, reflected in greater Bern score (P < .001) and increased expression of Sox9 (P = .013), Col2a1 (P < .001), and Acan (P < .001). Histologically, aged rats treated with M2-Exos demonstrated significantly greater histologic scores (P < .001 at both 6 and 12 weeks) and increased fibrocartilage regeneration at the enthesis. Biomechanically, these rats exhibited greater failure load, stiffness, and stress (all P < .001) at 12 weeks. Mechanistically, proteomic analysis suggested that M2-Exos might alleviate cellular senescence by potentially regulating DNA replication and repair. CONCLUSIONS M2-Exos can significantly alleviate BMSC senescence and thereby enhance tendon-to-bone healing in an aged rat RCT model. CLINICAL RELEVANCE This study suggests the potential utility of M2-Exos as a therapy for RCT in the older population.
Collapse
Affiliation(s)
- Zhuochang Cai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longqiang Shu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongyang Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuetao Xie
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xudong Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|