1
|
Gems D. How aging causes osteoarthritis: An evolutionary physiology perspective. Osteoarthritis Cartilage 2025:S1063-4584(25)01024-6. [PMID: 40381687 DOI: 10.1016/j.joca.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
Late-life diseases result from the poorly understood process of senescence (aging), which is largely genetically determined. According to a recently proposed evolutionary physiology-based account, the multifactorial model, senescence is largely caused by evolved but non-adaptive programmatic mechanisms specified by the wild-type (i.e. normal) genome. These act together with disruptions to wild-type function (due e.g. to infectious pathogens, mechanical injury and malnutrition) in a variety of combinations to generate diverse late-life diseases. Here, I explore the utility of this model by testing its capacity to provide an account of one complex, late-life disease, osteoarthritis (OA), and suggest a framework for understanding OA etiology. In this cartilage-focused framework, a core OA disease mechanism is a futile (non-adaptive) developmental program of endochondral ossification, in which hypertrophic articular cartilage chondrocytes alter joint architecture. Programmatic changes prime chondrocytes for futile program activation, which can be triggered by secondary causes of OA (e.g. joint mechanical injury). I suggest that an evolutionary cause of this priming, involving antagonistic pleiotropy, is selection to maximize early-life tissue repair benefits at the expense of late-life programmatic costs.
Collapse
Affiliation(s)
- David Gems
- Institute of Healthy Ageing, and Department of Genetics, Evolution and Environment, University College London, Gower Street, London, UK.
| |
Collapse
|
2
|
Li Y, Wang F, Ji B, Amati A, Cao L. FHL2 deteriorates IL-1β induced inflammation, apoptosis, and extracellular matrix degradation in chondrocyte-like ATDC5 cells by mTOR and NF-ĸB pathways. BMC Musculoskelet Disord 2025; 26:331. [PMID: 40186216 PMCID: PMC11971747 DOI: 10.1186/s12891-025-08536-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The role of nuclear translocation in osteoarthritis (OA) pathogenesis has garnered increasing attention in recent years. Extensive research has demonstrated that FHL2 acts as a nuclear transmitter through interactions with other nuclear transcription factors. We aimed to investigate the role of FHL2 in an osteoarthritis cell model. METHODS OA cartilage model was established by chondrocyte-like ATDC5 cells induced by 1% insulin-transferrin-selenium and then treated with interleukin-1β (IL-1β, 10 ng/mL). Lentivirus transfection was employed to suppress the expression of FHL2. Immunofluorescence and flow cytometry were used to examine nuclear transcription and apoptosis, respectively. Western blotting was performed to analyze the expression of metabolism-related proteins, autophagy-related proteins, apoptosis-related proteins, as well as proteins associated with the NF-ĸB and mTOR pathways. RESULTS The elevated expression of FHL2 occurred in both the cytoplasm and the nucleus. Knockdown of FHL2 could inhibit IL-1β-induced phosphorylation of NF-ĸB p65 and stabilize the extracellular matrix (ECM) by decreasing MMP-3 and MMP-13 expression, to suppress COL II degradation in chondrocyte-like ATDC5 cells. Meanwhile, the knockdown of FHL2-activated autophagy in IL-1β-treated chondrocytes through mTOR signaling, characterized by an increased LC3-II/LC3-I ratio and Beclin-1. FHL2 downregulation inhibited IL-1β-induced apoptosis by suppressing BAX and Caspase-3 expression, while enhancing BCL-2 protein levels. This mechanism may involve AKT phosphorylation and decreased expression of p-NF-ĸB p65. CONCLUSIONS FHL2 knockdown activated autophagy while suppressing inflammation, apoptosis, and ECM degradation. The mechanism underlying these processes may involve the inhibition of the mTOR and NF-ĸB signaling pathways.
Collapse
Affiliation(s)
- Yicheng Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University Urumqi, Xinjiang, P.R. China
| | - Fei Wang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University Urumqi, Xinjiang, P.R. China
| | - Baochao Ji
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University Urumqi, Xinjiang, P.R. China
| | - Abdusami Amati
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University Urumqi, Xinjiang, P.R. China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University Urumqi, Xinjiang, P.R. China.
| |
Collapse
|
3
|
Xu Y, Yang Y, Song H, Li M, Shi W, Yu T, Lin J, Yu Y. The Role of Exerkines in the Treatment of Knee Osteoarthritis: From Mechanisms to Exercise Strategies. Orthop Surg 2025; 17:1021-1035. [PMID: 39854050 PMCID: PMC11962297 DOI: 10.1111/os.14365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/25/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
With the increasing prevalence of knee osteoarthritis (KOA), the limitations of traditional treatments, such as their limited efficacy in halting disease progression and their potential side effects, are becoming more evident. This situation has prompted scientists to seek more effective strategies. In recent years, exercise therapy has gained prominence in KOA treatment due to its safety, efficacy, and cost-effectiveness, which are underpinned by the molecular actions of exerkines. Unlike conventional therapies, exerkines offer specific advantages by targeting inflammatory responses, enhancing chondrocyte proliferation, and slowing cartilage degradation at the molecular level. This review explores the potential mechanisms involved in and application prospects of exerkines in KOA treatment and provides a comprehensive analysis of their role. Studies show that appropriate exercise not only promotes overall health, but also positively impacts KOA by stimulating exerkine production. The effectiveness of exerkines, however, is influenced by exercise modality, intensity, and duration of exercise, making the development of personalized exercise plans crucial for KOA patients. Based on these insights, this paper proposes targeted exercise strategies designed to maximize exerkine benefits, aiming to provide novel perspectives for KOA prevention and treatment.
Collapse
Affiliation(s)
- Yuxiong Xu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Yizhuo Yang
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Hanan Song
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Ming Li
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Weihao Shi
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Tongwu Yu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| | - Jianhao Lin
- Arthritis Clinic & Research CenterPeking University People's HospitalBeijingChina
| | - Yanli Yu
- Sports & Medicine Integration Research CenterCapital University of Physical Education and SportsBeijingChina
| |
Collapse
|
4
|
Khadour FA, Khadour YA, Xu T. Electroacupuncture delays the progression of juvenile collagen-induced arthritis via regulation NLRP3/ NF-κB signaling pathway -mediated pyroptosis and its influence on autophagy. Clin Rheumatol 2025; 44:1713-1728. [PMID: 40067573 DOI: 10.1007/s10067-025-07354-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 04/13/2025]
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA) can lead to synovial inflammation. JIA is a chronic autoimmune inflammatory condition that primarily affects children. It is recognized as the most prevalent form of arthritis in the pediatric population and is associated with significant impairment and disability. Electroacupuncture (EA) effectively treats various synovium-related conditions, including symptoms of synovial inflammation, in both human and animal models. However, the specific mechanism by which EA protects against JIA remains unclear. Therefore, we conducted a comprehensive study to investigate the protective mechanisms of EA in a rat model. We aimed to examine the impact of EA on pathological changes in synovial tissue of juvenile collagen-induced arthritis (CIA) rats. METHODS The CIA model was established using Sprague‒Dawley (SD) rats aged 2-3 weeks. In this study, we investigated the potential role of EA on JIA by regulating the NLRP3-NF-κB axis in juvenile CIA rats and its influence on autophagy. To verify the effect of EA on juvenile CIA, the expression of NLRP3 was overexpressed by an adeno-associated virus injected into the knee joint of the CIA rats. RESULTS In this study, we observed that NLRP3 plays an important role in developing juvenile CIA and that NLRP3 overexpression exacerbates inflammation and increases synovium inflammation. We also demonstrated that the expression of NLRP3 was increased in synovial tissue, and NLRP3 could upregulate the NF-κB signal pathway and influence inflammation. Moreover, we also found increases in the expression of NLRP3 by impairing autophagy capacity and activation of the pyroptosis pathway in the synovium of the juvenile CIA rats. CONCLUSION Moreover, we also discovered that EA decreased the expression of NLRP3 by restoring the impaired autophagy capacity and inhibiting the NLRP3-NF-κB axis, thereby delaying the progression of juvenile CIA. These results showed that EA is effective in inhibiting inflammation and synovial degeneration and alleviating the progression of juvenile CIA. As a result, our results provide new insight into the mechanism by which EA delays the development of juvenile CIA, offering a novel therapeutic regimen for JIA. This trial was registered with ClinicalTrials.gov, number NCT10203935. Registered October 07, 2023. Key Points • NLRP3 plays a critical role in juvenile collagen-induced arthritis (CIA), with its overexpression linked to increased inflammation in synovial tissue. • Electroacupuncture (EA) reduces NLRP3 expression and inhibits the NLRP3-NF-κB axis, mitigating inflammatory responses and delaying juvenile CIA progression. • EA restores impaired autophagy in juvenile CIA rats, promoting cellular health and inflammation management. • EA alleviates synovial degeneration, improving joint health and function in juvenile CIA models.
Collapse
Affiliation(s)
- Fater A Khadour
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Rehabilitation, Faculty of Medicine, Al Baath University, Homs, Syria
- Department of Physical Therapy, Health Science Faculty, Al-Baath University, Homs, Syria
| | - Younes A Khadour
- Department of Rehabilitation, Faculty of Medicine, Al Baath University, Homs, Syria
- Department of Physical Therapy, Cairo University, Cairo, 11835, Egypt
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
5
|
Guan M, Han X, Liao B, Han W, Chen L, Zhang B, Peng X, Tian Y, Xiao G, Li X, Kuang L, Zhu Y, Bai D. LIPUS Promotes Calcium Oscillation and Enhances Calcium Dependent Autophagy of Chondrocytes to Alleviate Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413930. [PMID: 40013941 PMCID: PMC12021083 DOI: 10.1002/advs.202413930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/02/2025] [Indexed: 02/28/2025]
Abstract
Osteoarthritis (OA) is a degenerative disease which places an enormous burden on society, effective treatments are still limited. As a non-invasive and safe physical therapy, low-intensity pulsed ultrasound (LIPUS) can alleviate OA progression, but the underlying mechanism is not fully understood, especially the mechanical transduction between LIPUS and the organism. In this pioneering study, the biomechanical effects of LIPUS on living mice chondrocytes and living body zebrafish are investigate by using fluorescence imaging technology, to dynamically "visualize" its invisible mechanical stimuli in the form of calcium oscillations. It is also confirmed that LIPUS maintains cartilage homeostasis by promoting chondrocyte autophagy in a calcium-dependent manner. In addition, chondrocyte ion channels are screened by scRNA-seq and confirm that the mechanosensitive ion channel transient receptor potential vanilloid 4 (TRPV4) mediated the biological effects of LIPUS on chondrocytes. Finally, it is found that a combination of pharmacologically induced and LIPUS-induced Ca2+ influx in chondrocytes enhances the cartilage-protective effect of LIPUS, which may provide new insights for optimizing LIPUS in the treatment of OA.
Collapse
Affiliation(s)
- Mengtong Guan
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaoyu Han
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqing400016China
| | - Bo Liao
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Wang Han
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Lin Chen
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Bin Zhang
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Xiuqin Peng
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Yu Tian
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Gongyi Xiao
- Department of OrthopedicsChonggang General HospitalChongqing400000China
| | - Xinhe Li
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqing400016China
| | - Liang Kuang
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Ying Zhu
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Dingqun Bai
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqing400016China
| |
Collapse
|
6
|
Lin M, Zhang C, Li H, Li K, Gou S, He X, Lv C, Gao K. Pyroptosis for osteoarthritis treatment: insights into cellular and molecular interactions inflammatory. Front Immunol 2025; 16:1556990. [PMID: 40236711 PMCID: PMC11996656 DOI: 10.3389/fimmu.2025.1556990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Osteoarthritis (OA) is a widely prevalent chronic degenerative disease often associated with significant pain and disability. It is characterized by the deterioration of cartilage and the extracellular matrix (ECM), synovial inflammation, and subchondral bone remodeling. Recent studies have highlighted pyroptosis-a form of programmed cell death triggered by the inflammasome-as a key factor in sustaining chronic inflammation. Central to this process are the inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18), which play crucial roles mediating intra-articular pyroptosis through the NOD-like receptor protein 3 (NLRP3) inflammasome. This paper investigates the role of the pyroptosis pathway in perpetuating chronic inflammatory diseases and its linkage with OA. Furthermore, it explores the mechanisms of pyroptosis, mediated by nuclear factor κB (NF-κB), the purinergic receptor P2X ligand-gated ion channel 7 (P2X7R), adenosine monophosphate (AMP)-activated protein kinase (AMPK), and hypoxia-inducible factor-1α (HIF-1α). Additionally, it examines the interactions among various cellular components in the context of OA. These insights indicate that targeting the regulation of pyroptosis presents a promising therapeutic approach for the prevention and treatment of OA, offering valuable theoretical perspectives for its effective management.
Collapse
Affiliation(s)
- Minghui Lin
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cunxin Zhang
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Haiming Li
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kang Li
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Shuao Gou
- Jining No.1 People's Hospital, affiliated with Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao He
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
- Medical Integration and Practice Center, Shandong University, Jinan, China
| | - Chaoliang Lv
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Kai Gao
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
7
|
Li M, Wei CB, Li HF, He K, Bai RJ, Zhang FJ. Osteopontin inhibits autophagy via CD44 and avβ3 integrin and promotes cell proliferation in osteoarthritic fibroblast-like synoviocytes. BMC Musculoskelet Disord 2025; 26:274. [PMID: 40102843 PMCID: PMC11916941 DOI: 10.1186/s12891-025-08509-y] [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: 08/09/2023] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is closely related to aging, and autophagy is implicated in the retardation of aging. Activated synoviocytes play important roles in OA; the synoviocytes could produce osteopontin (OPN) and its main receptors CD44 and integrin, which are all involved in OA. The purpose of this study is to investigate whether OPN has an effect on autophagy in osteoarthritic synoviocytes. METHODS We cultured human OA fibroblast-like synoviocytes (FLS) and treated them with rhOPN and antibodies against CD44 and CD51/61 (αvβ3 integrin) or isotype IgG to block the interaction between receptors and ligands. Infection with lentivirus mRFP-GFP-LC3, laser confocal imaging and Western blotting were used to determine changes in the expression of autophagy markers, and cell proliferation of FLS was assessed with a CCK-8 assay. RESULTS Our results showed the expression level of autophagy marker protein LC3 II and the mRFP-GFP-LC3 puncta were significantly decreased after treatment with rhOPN when compared with the control group, when the FLS were incubated with antibodies against CD44 or CD51/61 (αvβ3 integrin) or with control isotype IgG for 1 h, followed by rhOPN treatment for 48 h, rhOPN could suppress the relative expression of LC3 II and Beclin1 via integrin and CD44 in the FLS, CCK-8 assay also showed that rhOPN significantly increased the cell proliferation and viability of FLS. CONCLUSIONS OPN could inhibit autophagy via CD44 and αvβ3 integrin and promote the proliferation of FLS, playing an important role in OA synovitis.
Collapse
Affiliation(s)
- Min Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Chang-Bao Wei
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Hai-Feng Li
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
| | - Rui-Jun Bai
- Department of Orthopaedics, Wuxi Ninth People's Hospital, Soochow University, 999 Liangxi Road, Wuxi, Jiangsu, 214000, China.
| | - Fang-Jie Zhang
- Department of Emergency Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), No.87 Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
8
|
Deng Y, Hou M, Wu Y, Liu Y, Xia X, Yu C, Yu J, Yang H, Zhang Y, Zhu X. SIRT3-PINK1-PKM2 axis prevents osteoarthritis via mitochondrial renewal and metabolic switch. Bone Res 2025; 13:36. [PMID: 40087281 PMCID: PMC11909255 DOI: 10.1038/s41413-025-00413-4] [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] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 03/17/2025] Open
Abstract
Maintaining mitochondrial homeostasis is critical for preserving chondrocyte physiological conditions and increasing resistance against osteoarthritis (OA). However, the underlying mechanisms governing mitochondrial self-renewal and energy production remain elusive. In this study, we demonstrated mitochondrial damage and aberrant mitophagy in OA chondrocytes. Genetically overexpressing PTEN-induced putative kinase 1 (PINK1) protects against cartilage degeneration by removing defective mitochondria. PINK1 knockout aggravated cartilage damage due to impaired mitophagy. SIRT3 directly deacetylated PINK1 to promote mitophagy and cartilage anabolism. Specifically, PINK1 phosphorylated PKM2 at the Ser127 site, preserving its active tetrameric form. This inhibited nuclear translocation and the interaction with β-catenin, resulting in a metabolic shift and increased energy production. Finally, a double-knockout mouse model demonstrated the role of the SIRT3-PINK1-PKM2 axis in safeguarding the structural integrity of articular joints and improving motor functions. Overall, this study provides a novel insight into the regulation of mitochondrial renewal and metabolic switches in OA.
Collapse
Affiliation(s)
- Yaoge Deng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Yubin Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Xiaowei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Chenqi Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Jianfeng Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China.
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China.
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
9
|
Nogueira-Recalde U, Lambertucci F, Montégut L, Motiño O, Chen H, Lachkar S, Anagnostopoulos G, Stoll G, Li S, Carbonier V, Saavedra Díaz E, Blanco FJ, van Tetering G, de Boer M, Maiuri MC, Caramés B, Martins I, Kroemer G. Neutralization of acyl CoA binding protein (ACBP) for the experimental treatment of osteoarthritis. Cell Death Differ 2025:10.1038/s41418-025-01474-y. [PMID: 40082721 DOI: 10.1038/s41418-025-01474-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
The plasma concentrations of acyl CoA binding protein (ACBP) encoded by the gene diazepam binding inhibitor (DBI) are increased in patients with severe osteoarthritis (OA). Here, we show that knee OA induces a surge in plasma ACBP/DBI in mice subjected to surgical destabilization of one hind limb. Knockout of the Dbi gene or intraperitoneal (i.p.) injection of a monoclonal antibody (mAb) neutralizing ACBP/DBI attenuates OA progression in this model, supporting a pathogenic role for ACBP/DBI in OA. Furthermore, anti-ACBP/DBI mAb was also effective against OA after its intraarticular (i.a.) injection, as monitored by sonography, revealing the capacity of ACBP/DBI to locally reduce knee inflammation over time. In addition, i.a. anti-ACBP/DBI mAb improved functional outcomes, as indicated by the reduced weight imbalance caused by OA. At the anatomopathological level, i.a. anti-ACBP/DBI mAb mitigated histological signs of joint destruction and synovial inflammation. Of note, i.a. anti-ACBP/DBI mAb blunted the OA-induced surge of plasma ACBP/DBI, as well as that of other inflammatory factors including interleukin-1α, interleukin-33, and tumor necrosis factor. These findings are potentially translatable to OA patients because joints from OA patients express both ACBP/DBI and its receptor GABAARγ2. Moreover, a novel mAb against ACBP/DBI recognizing an epitope conserved between human and mouse ACBP/DBI demonstrated similar efficacy in mitigating OA as an anti-mouse ACBP/DBI-only mAb. In conclusion, ACBP/DBI might constitute a promising therapeutic target for the treatment of OA.
Collapse
Affiliation(s)
- Uxía Nogueira-Recalde
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Unidad de Biología del Cartílago, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidad de A Coruña (UDC), A Coruña, Spain
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Omar Motiño
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Unidad de Excelencia, Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - CSIC, Valladolid, Spain
| | - Hui Chen
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Sylvie Lachkar
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Gerasimos Anagnostopoulos
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Gautier Stoll
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Sijing Li
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Vincent Carbonier
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Ester Saavedra Díaz
- Departamento de Bioquímica y Biología Molecular, Fisiología, Genética e Inmunología, Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Francisco J Blanco
- Unidad de Biología del Cartílago, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidad de A Coruña (UDC), A Coruña, Spain
| | | | | | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Beatriz Caramés
- Unidad de Biología del Cartílago, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidad de A Coruña (UDC), A Coruña, Spain
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France.
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
10
|
Minton DM, Ailiani AR, Focht MD, Kersh ME, Li A, Elliehausen CJ, Sonsalla MM, Lamming DW, Marolf AJ, Santangelo KS, Salmon AB, Konopka AR. Inhibition of mTORC1 by rapamycin results in feedback activation of Akt S473 and aggravates hallmarks of osteoarthritis in female mice and non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.14.594256. [PMID: 38798488 PMCID: PMC11118493 DOI: 10.1101/2024.05.14.594256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Purpose Genetic deletion of mTOR has protected against post-traumatic osteoarthritis (OA) in male mice, however, effects of pharmacological mTOR-inhibition are equivocal and have not been tested in aging models nor in female subjects. Therefore, the goal of this study was to determine if mTOR-inhibition by rapamycin can modify OA pathology in aging non-human primates and female mice. Methods Common marmosets were administered oral rapamycin (1mg/kg/day) or vehicle starting near mid-life until death. Five-month-old, female C57BL/6J mice were treated with vehicle or rapamycin (IP, 2mg/kg, 3x/week) for 8-weeks following non-invasive ACL rupture. Knee OA pathology was assessed via microCT and histology. Phosphorylation of mTORC1 (p-RPS6S235/36) and mTORC2 (p-AktS473, p-NDRG1T638, p-PKCαT348) substrates were evaluated via western blot in articular cartilage, meniscus, and/or infrapatellar fat pad. ATDC5 cells were cultured with rapamycin to determine time and dose effects on mTORC1/2 signaling. Results In marmosets, rapamycin did not impact age-related radiographic OA severity or cartilage pathology but increased medial meniscus calcification and lowered lateral tibia subchondral thickness, particularly in females. In female mice, rapamycin worsened ACLR-induced meniscus calcification and cartilage pathology. In marmoset and mouse joint tissues, rapamycin inhibited mTORC1 and increased p-AktS473 but not p-NDRG1T638 or p-PKCαT348. This mTOR signaling pattern was replicated in ATDC5 cells during exposure to low concentrations of rapamycin. Conclusions Rapamycin attenuated mTORC1 signaling with feedback activation of AktS473 in articular cartilage, meniscus, and/or infrapatellar fat pad and was accompanied by deleterious effects on meniscus calcification and/or cartilage pathology in female mice and common marmosets.
Collapse
Affiliation(s)
- Dennis M. Minton
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Aditya R. Ailiani
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Michael D.K. Focht
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Mariana E. Kersh
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Champaign, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Amy Li
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Christian J. Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Michelle M. Sonsalla
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Dudley W. Lamming
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Angela J. Marolf
- Department of Veterinary Clinical Sciences, Ohio State University, Columbus, OH, USA
| | - Kelly S Santangelo
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Adam B. Salmon
- Barshop Institute for Longevity and Aging Studies and Departments of Molecular Medicine and Cellular and Integrative Physiology, University of Texas Health Science Center, San Antonio, TX, USA
- Geriatric Research, Education, and Clinical Center, South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - Adam R. Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
11
|
Khadour FA, Khadour YA, Xu T. NLRP3 overexpression exacerbated synovium tissue degeneration in juvenile collagen-induced arthritis. Sci Rep 2025; 15:7024. [PMID: 40016261 PMCID: PMC11868420 DOI: 10.1038/s41598-025-86720-6] [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] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/13/2025] [Indexed: 03/01/2025] Open
Abstract
Juvenile idiopathic arthritis (JIA) can lead to synovial inflammation. JIA is a chronic autoimmune inflammatory condition that primarily affects children. It is recognized as the most prevalent form of arthritis in the pediatric population and is associated with significant impairment and disability. As an inflammatory regulator, Nod-like receptor 3 (NLRP3) has been implicated in various autoimmune diseases. However, the specific mechanism by which NLRP3 impacts the progress of JIA remains unclear. Therefore, we conducted this study to investigate the specific mechanism of NLRP3 on the progress of synovial inflammation in juvenile collagen-induced arthritis (CIA). The CIA model was established using Sprague‒Dawley (SD) rats aged 2-3 weeks. In this study, we investigated the potential role of NLRP3 on JIA by regulating the NLRP3-NF-κB axis in CIA rats. To verify the effect of NLRP3 on JIA, the expression of NLRP3 was knocked down or overexpressed by an adeno-associated virus injected into the knee joint of the CIA rats. In this study, we observed that NLRP3 plays an important role in the development of juvenile CIA, and knocking down NLRP3 inhibited inflammation and alleviated synovium inflammation. We also demonstrated that the expression of NLRP3 was increased in synovial tissue, and NLRP3 could upregulate the NF-κB signal pathway and influence inflammation. Moreover, we also found that increases in the expression of NLRP3 impairs autophagy capacity and increases activation of the pyroptosis pathway in the synovium of the juvenile CIA rats. The results demonstrated that NLRP3 interferes with synovial inflammation in juvenile CIA. These results provide new insight into the mechanism by which NLRP3 impacts the development of JIA and suggest that targeting the NLRP3 inflammasome may represent a promising therapeutic strategy for managing JIA.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Rats
- Synovial Membrane/pathology
- Synovial Membrane/metabolism
- Rats, Sprague-Dawley
- Arthritis, Experimental/pathology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/genetics
- NF-kappa B/metabolism
- Arthritis, Juvenile/pathology
- Arthritis, Juvenile/metabolism
- Arthritis, Juvenile/genetics
- Male
- Signal Transduction
- Disease Models, Animal
- Inflammasomes/metabolism
Collapse
Affiliation(s)
- Fater A Khadour
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
- Department of Rehabilitation, Faculty of Medicine, Al Baath University, Homs, Syria
- Department of Physical Therapy, Health Science Faculty, Al-Baath University, Homs, Syria
| | - Younes A Khadour
- Department of Rehabilitation, Faculty of Medicine, Al Baath University, Homs, Syria
- Department of Physical Therapy, Cairo University, Cairo, 11835, Egypt
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China.
| |
Collapse
|
12
|
Morici L, Allémann E, Jordan O, Nikolić I. Promising LOX proteins for cartilage-targeting osteoarthritis therapy. Pharmacol Res 2025; 212:107627. [PMID: 39875019 DOI: 10.1016/j.phrs.2025.107627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
Osteoarthritis (OA) is the most affected joint disease worldwide, touching millions of people every year. It is caused by a progressive degeneration of articular cartilage, causing pain and limited mobility. Among the pathways involved in cartilage homeostasis, "LOX" proteins (referring to three distinct protein families, very often confused in the literature) play a prominent role. The lipoxygenase enzyme family is involved in the inflammatory process of OA by inducing the production of several pro-inflammatory leukotrienes. Lectin-like oxidized low-density lipoprotein family are receptors located at the surface of chondrocytes, which interact with their ligand, ox-LDL, activating several catabolic pathways involved in OA pathophysiology. Finally, lysyl oxidase and lysyl oxidase-like are enzymes expressed intracellularly (in chondrocytes' cytoplasm) involved in elastin biosynthesis and collagen cross-linking in cartilage extracellular matrix. EMA and FDA have not yet approved any drug targeting the LOX proteins. In particular, today lysyl oxidase-like 2 is considered as a new promising target for OA modifying therapy. This review clarifies the main roles of different LOX proteins involved in the progression of OA. Potential LOX inhibitoion strategies for drug development in advanced OA therapy, particularly for local intraarticular delivery, were listed and discussed for each target type. This review, therefore, proposes promising strategies for future drug development in OA treatment.
Collapse
Affiliation(s)
- Luca Morici
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland
| | - Ines Nikolić
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, Geneva 4, 1211, Switzerland.
| |
Collapse
|
13
|
Xu H, Luo Y, An Y, Wu X. The mechanism of action of indole-3-propionic acid on bone metabolism. Food Funct 2025; 16:406-421. [PMID: 39764708 DOI: 10.1039/d4fo03783a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Indole-3-propionic acid (IPA), a metabolite produced by gut microbiota through tryptophan metabolism, has recently been identified as playing a pivotal role in bone metabolism. IPA promotes osteoblast differentiation by upregulating mitochondrial transcription factor A (Tfam), contributing to increased bone density and supporting bone repair. Simultaneously, it inhibits the formation and activity of osteoclasts, reducing bone resorption, possibly through modulation of the nuclear factor-κB (NF-κB) pathway and downregulation of osteoclast-associated factors, thereby maintaining bone structural integrity. Additionally, IPA provides indirect protection to bone health by regulating host immune responses and inflammation via activation of receptors such as the Aryl hydrocarbon Receptor (AhR) and the Pregnane X Receptor (PXR). This review summarizes the roles and signaling pathways of IPA in bone metabolism and its impact on various bone metabolic disorders. Furthermore, we discuss the therapeutic potential and limitations of IPA in treating bone metabolic diseases, aiming to offer novel strategies for clinical management.
Collapse
Affiliation(s)
- Huimin Xu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi An
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Xi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Xie X, Chao R, Mao Y, Wan T, Wang Y, Zhu Y, Xu W, Chen X, Wang Y, Ma Z, Zhang S. Osteoarthritis-like changes in rat temporomandibular joint induced by unilateral anterior large overjet treatment. Sci Rep 2025; 15:1646. [PMID: 39794380 PMCID: PMC11723919 DOI: 10.1038/s41598-024-81306-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a common degenerative disease that causes chronic pain and joint dysfunction. However, the current understanding of TMJOA pathogenesis is limited and necessitates further research. Animal models are crucial for investigating TMJOA due to the scarcity of clinical samples. Class II malocclusion is an occlusal type highly associated with TMJOA, but it currently lacks appropriate animal models for simulating this malocclusion in research. Therefore, this study develops a new malocclusion model using a unilateral anterior large overjet (UALO) dental device to cause Class II malocclusion characteristics and TMJOA-like pathological alterations in rats. By inducing a posteriorly positioned condyle, the UALO device effectively results in cartilage degradation, subchondral bone loss, condylar volume reduction, and mandibular retrusion. Furthermore, RNA sequencing of condylar cartilages revealed that the oxidative stress of chondrocytes was elevated under the UALO-triggered abnormal mechanical stress. Disruption of antioxidant systems and mitochondrial dysfunction are involved in cartilage degeneration. The current study provides a novel and reliable rat model suitable for TMJOA research and offers insights into the disease's potential mechanistic pathways and molecular targets, contributing to a deeper understanding of TMJOA.
Collapse
Affiliation(s)
- Xinru Xie
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Rui Chao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yi Mao
- State Key Laboratory of Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Tianhao Wan
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yexin Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yan Zhu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Weifeng Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Xuzhuo Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yong Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Zhigui Ma
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Shanyong Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
15
|
Florencio-Silva R, Sasso GRDS, Sasso-Cerri E, Cerri PS, Gil CD, de Jesus Simões M. Relationship between autophagy and NLRP3 inflammasome during articular cartilage degradation in oestrogen-deficient rats with streptozotocin-induced diabetes. Ann Anat 2025; 257:152318. [PMID: 39216675 DOI: 10.1016/j.aanat.2024.152318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Estrogen deficiency and Diabetes mellitus (DM) cause joint tissue deterioration, although the mechanisms are uncertain. This study evaluated the immunoexpression of autophagy and NLRP3-inflammasome markers, in rat articular cartilage with estrogen deficiency and DM. METHODS Twenty rats were sham-operated (SHAM) or ovariectomized (OVX) and equally allocated into four groups: SHAM and OVX groups administered with vehicle solution; SHAM and OVX groups treated with 60 mg/kg/body weight of streptozotocin, intraperitoneally, to induce DM (SHAM-DM and OVX-DM groups). After seven weeks, the rats were euthanized, and their joint knees were processed for paraffin embedding. Sections were stained with haematoxylin-eosin, toluidine blue, safranin-O/fast-green or subjected to picrosirius-red-polarisation method; immunohistochemistry to detect beclin-1 and microtubule-associated protein 1B-light chain 3 (autophagy markers), NLRP3 and interleukin-1β (IL-1β) (inflammasome activation markers), along with matrix metalloproteinase-9 (MMP-9), Nuclear factor-kappa B (NFκB), and Vascular endothelial growth factor A (VEGF-A) were performed. RESULTS Deterioration of articular cartilage and subchondral bone were greater in SHAM-DM and OVX-DM groups. Higher percentages of immunolabeled chondrocytes to NLRP3, IL-1β, MMP-9, NFκB, and VEGF-A, as well as lower percentages of chondrocytes immunolabeled to autophagy markers, were noticed in estrogen-deficient and diabetic groups. These differences were greater in the OVX-DM group. Percentages of immunolabeled chondrocytes showed negative correlation between autophagy markers v.s IL-1β, NLRP-3, MMP-9, NFκB, and VEGF-A, along with positive correlation between VEGF-A vs. MMP-9, NFκB, IL-1β, and NLRP3, and MMP-9 vs. NFκB. CONCLUSIONS In conclusion, autophagy reduction and NLRP3 inflammasome activation in chondrocytes may be implicated in articular cartilage degradation, under estrogen-deficient and DM conditions. Moreover, the combination of estrogen deficiency and DM may potentiate those effects.
Collapse
Affiliation(s)
- Rinaldo Florencio-Silva
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil.
| | - Gisela Rodrigues da Silva Sasso
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Paulo Sérgio Cerri
- São Paulo State University (UNESP), School of Dentistry, Araraquara - Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry - Laboratory of Histology and Embryology, Araraquara, SP, Brazil
| | - Cristiane Damas Gil
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| | - Manuel de Jesus Simões
- Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Ginecologia, São Paulo, SP, Brazil; Universidade Federal de São Paulo - UNIFESP, Escola Paulista de Medicina - EPM, Departamento de Morfologia e Genética, Disciplina de Histologia e Biologia Estrutural, São Paulo, SP, Brazil
| |
Collapse
|
16
|
Zhang Z, He T, Gu H, Zhao Y, Tang S, Han K, Hu Y, Wang H, Yu H. Single-cell RNA sequencing identifies the expression of hemoglobin in chondrocyte cell subpopulations in osteoarthritis. BMC Mol Cell Biol 2024; 25:28. [PMID: 39736555 DOI: 10.1186/s12860-024-00519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/02/2024] [Indexed: 01/01/2025] Open
Abstract
In recent years, chondrocytes have been found to contain hemoglobin, which might be an alternative strategy for adapting to the hypoxic environment, while the potential mechanisms of that is still unclear. Here, we report the expression characteristics and potential associated pathways of hemoglobin in chondrocytes using single-cell RNA sequencing (scRNA-seq). We downloaded data of normal people and patients with osteoarthritis (OA) from the Gene Expression Omnibus (GEO) database and cells are unbiased clustered based on gene expression pattern. We determined the expression levels of hemoglobin in various chondrocyte subpopulations. Meanwhile, we further explored the difference in the enriched signaling pathways and the cell-cell interaction in chondrocytes of the hemoglobin high-expression and low-expression groups. Specifically, we found that SPP1 was closely associated with the expression of hemoglobin in OA progression. Our findings provide new insights into the distribution characteristics of hemoglobin in chondrocytes and provide potential clues to the underlying role of hemoglobin in OA and the mechanisms related to that, providing potential new ideas for the treatment of OA.
Collapse
Affiliation(s)
- Zhihao Zhang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Ting He
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongwen Gu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Yuanhang Zhao
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Shilei Tang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Kangen Han
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Yin Hu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Hongwei Wang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China.
| | - Hailong Yu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China.
| |
Collapse
|
17
|
Andrei C, Mihai DP, Nitulescu GM, Nitulescu G, Zanfirescu A. Modulating Autophagy in Osteoarthritis: Exploring Emerging Therapeutic Drug Targets. Int J Mol Sci 2024; 25:13695. [PMID: 39769455 PMCID: PMC11727697 DOI: 10.3390/ijms252413695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by the breakdown of cartilage and the subsequent inflammation of joint tissues, leading to pain and reduced mobility. Despite advancements in symptomatic treatments, disease-modifying therapies for OA remain limited. This narrative review examines the dual role of autophagy in OA, emphasizing its protective functions during the early stages and its potential to contribute to cartilage degeneration in later stages. By delving into the molecular pathways that regulate autophagy, this review highlights its intricate interplay with oxidative stress and inflammation, key drivers of OA progression. Emerging therapeutic strategies aimed at modulating autophagy are explored, including pharmacological agents such as AMP kinase activators, and microRNA-based therapies. Preclinical studies reveal encouraging results, demonstrating that enhancing autophagy can reduce inflammation and decelerate cartilage degradation. However, the therapeutic benefits of autophagy modulation depend on precise, stage-specific approaches. Excessive or dysregulated autophagy in advanced OA may lead to chondrocyte apoptosis, exacerbating joint damage. This review underscores the promise of autophagy-based interventions in bridging the gap between experimental research and clinical application. By advancing our understanding of autophagy's role in OA, these findings pave the way for innovative and effective therapies. Nonetheless, further research is essential to optimize these strategies, address potential off-target effects, and develop safe, targeted treatments that improve outcomes for OA patients.
Collapse
Affiliation(s)
| | - Dragos Paul Mihai
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.A.); (G.M.N.); (G.N.); (A.Z.)
| | | | | | | |
Collapse
|
18
|
Chen Z, Tang M, Wu Z, Lin Y, Wu C, Huang H, Chen J, Zhu Z, Liu Y, Tang S, Ding C, Han W. Increased Rab1a accelerates osteoarthritis by inhibiting autophagy via activation of the mTORC1-S6K pathway. J Adv Res 2024:S2090-1232(24)00501-0. [PMID: 39521431 DOI: 10.1016/j.jare.2024.11.009] [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: 07/09/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Cartilage degradation is a critical alteration in the progression of osteoarthritis (OA) due to the disorder of chondrocyte metabolic homeostasis. Autophagy plays an important role in maintaining intracellular homeostasis. Recent investigations have increasingly underscored the importance of autophagy in modulating the pathological mechanisms underlying OA. Ras-related protein Rab-1a (Rab1a) has been illustrated to regulate autophagy in many diseases but not in OA. OBJECTIVES This study aims to elucidate whether Rab1a could regulate the development of OA through modulation of chondrocyte autophagy and apoptosis. METHODS Proteomic sequencing, Western blotting, and immunohistochemistry were applied to detect the expression level of Rab1a in vitro and in vivo. Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways were rigorously identified. The effects of Rab1a and the interaction between Rab1a, mTORC1, autophagy and apoptosis were explored by qPCR, Western blotting, and immunofluorescence. An experimental mouse OA model was also performed to confirm the role of Rab1a in OA pathogenesis in vivo. Histological analysis was employed to demonstrate cartilage damage. RESULTS Rab1a expression was significantly upregulated in inflamed chondrocytes and knee OA cartilage. Inhibition of Rab1a partially attenuated the degradation of the extracellular matrix and cell apoptosis both in vitro and in vivo, whereas overexpression of Rab1a intensified cartilage matrix degradation and cellular apoptosis. Additionally, elevated Rab1a levels were observed to suppress autophagy and activate the mTORC1-S6K signaling pathway, thereby aggravating OA pathogenesis. CONCLUSION The augmentation of Rab1a expression impairs autophagy and promotes apoptosis through the activation of the mTORC1-S6K signaling pathway, further exacerbating OA pathogenesis. This finding suggests that Rab1a serves as a promising and innovative therapeutic target for the prevention and treatment of OA.
Collapse
Affiliation(s)
- Ze Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, The Seventh Affiliated Hospital, Southern Medical University, 28 Liguan Road, Nanhai District, Foshan, Guangdong 528244, China
| | - Mingze Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zewei Wu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yongcong Lin
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Cuixi Wu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hong Huang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jianmao Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Yongming Liu
- Centre of Orthopedics, The Seventh Affiliated Hospital, Southern Medical University, 28 Liguan Road, Nanhai District, Foshan, Guangdong 528244, China
| | - Súan Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Department of Spinal Surgery, Orthopedicdical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Institute of Exercise and Rehabilitation Science, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China.
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia.
| | - Weiyu Han
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Centre of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China.
| |
Collapse
|
19
|
Dhanabalan KM, Padhan B, Dravid AA, Agarwal S, Pancheri NM, Lin A, Willet NJ, Padmanabhan AK, Agarwal R. Nordihydroguaiaretic acid microparticles are effective in the treatment of osteoarthritis. J Mater Chem B 2024; 12:11172-11186. [PMID: 39356214 DOI: 10.1039/d4tb01342e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Several disease-modifying osteoarthritis (OA) drugs have emerged, but none have been approved for clinical use due to their systemic side effects, short half-life, and rapid clearance from the joints. Nordihydroguaiaretic acid (NDGA), a reactive oxygen species (ROS) scavenger and autophagy inducer, could be a potential treatment for OA. In this report, we show for the first time that sustained delivery of NDGA through polymeric microparticles maintains therapeutic concentrations of drug in the joint and ameliorates post-traumatic OA (PTOA) in a mouse model. In vitro treatment of oxidatively stressed primary chondrocytes from OA patients using NDGA-loaded poly(lactic-co-glycolic acid) (PLGA) microparticles (NDGA-MP) inhibited 15-lipoxygenase, induced autophagy, prevented chondrosenescence, and sustained matrix production. In vivo intra-articular delivery of NDGA-MP was non-toxic and had prolonged retention time (up to 35 days) in murine knee joints. Intra-articular therapy using NDGA-MP effectively reduced cartilage damage and reduced pain in the surgery-induced PTOA mouse model. Our studies open new avenues to modulate the immune environment and treat post-traumatic OA using ROS quenchers and autophagy inducers.
Collapse
Affiliation(s)
- Kaamini M Dhanabalan
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Bhagyashree Padhan
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Ameya A Dravid
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Smriti Agarwal
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| | - Nicholas M Pancheri
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, OR, USA, 97403
| | - Angela Lin
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, OR, USA, 97403
| | - Nick J Willet
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, OR, USA, 97403
| | | | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India, 560012.
| |
Collapse
|
20
|
Zou Y, Wang Z, Shi H, Hu J, Hu W. Soybean Isoflavones Alleviate Osteoarthritis Through Modulation of the TSC1/mTORC1 Signaling Pathway to Reduce Intrachondral Angiogenesis. Immunol Invest 2024; 53:1439-1455. [PMID: 39360672 DOI: 10.1080/08820139.2024.2410737] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
BACKGROUND The incidence of osteoarthritis (OA) is increasing, yet its pathogenesis remains largely unknown. Recent studies suggest that abnormal subchondral bone remodeling plays a crucial role in OA development, highlighting a gap in clinical treatments targeting this aspect. Soybean Isoflavone (SI) has shown potential in treating OA, although its mechanisms are not fully understood. METHODS This research investigated the effects of SI on subchondral bone remodeling in an OA rat model, assessing joint damage, OARSI scores, and type H vessel formation (CD31hiEmcnhi expression). Additionally, the expression of ALP, OCN, BMP, and TSC1 was evaluated to determine involvement of the mTORC1 pathway. In vitro studies on IL-1β-induced osteoblasts further examined the impact of SI on TSC1/mTORC1 signaling and related markers. RESULTS SI treatment reduced joint damage and OARSI scores in the rat OA model, significantly decreasing CD31hiEmcnhi expression, indicating a reduction in type H vessel formation. SI also downregulated ALP, OCN, and BMP expression while upregulating TSC1, suggesting inhibition of the mTORC1 signaling pathway and VEGF release. In vitro, SI increased TSC1 expression and decreased mTORC1 signaling, VEGF, ALP, OCN, and BMP levels in IL-1β-induced osteoblasts. CONCLUSION SI targets the TSC1/mTORC1 signaling pathway to suppress osteoblast activation and VEGF release, inhibiting type H vessel formation and slowing abnormal subchondral bone remodeling. These findings provide a novel therapeutic approach for OA by focusing on subchondral bone remodeling mechanisms.
Collapse
Affiliation(s)
- Yang Zou
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaoyang Wang
- Department of Orthopedics, Chengdu Bayi Orthopedic Hospital, Chengdu, China
| | - Hangchu Shi
- Department of Orthopedics, The Third People's Hospital of Yuhang District, Hangzhou, China
| | - Jiong Hu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Hu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
21
|
Li L, Li J, Li JJ, Zhou H, Zhu XW, Zhang PH, Huang B, Zhao WT, Zhao XF, Chen ES. Chondrocyte autophagy mechanism and therapeutic prospects in osteoarthritis. Front Cell Dev Biol 2024; 12:1472613. [PMID: 39507422 PMCID: PMC11537998 DOI: 10.3389/fcell.2024.1472613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis characterized by progressive cartilage degradation, with its pathogenesis closely related to chondrocyte autophagy. Chondrocytes are the only cells in articular cartilage, and the function of chondrocytes plays a vital role in maintaining articular cartilage homeostasis. Autophagy, an intracellular degradation system that regulates energy metabolism in cells, plays an incredibly important role in OA. During the early stages of OA, autophagy is enhanced in chondrocytes, acting as an adaptive mechanism to protect them from various environmental changes. However, with the progress of OA, chondrocyte autophagy gradually decreases, leading to the accumulation of damaged organelles and macromolecules within the cell, prompting chondrocyte apoptosis. Numerous studies have shown that cartilage degradation is influenced by the senescence and apoptosis of chondrocytes, which are associated with reduced autophagy. The relationship between autophagy, senescence, and apoptosis is complex. While autophagy is generally believed to inhibit cellular senescence and apoptosis to promote cell survival, recent studies have shown that some proteins are degraded by selective autophagy, leading to the secretion of the senescence-associated secretory phenotype (SASP) or increased SA-β-Gal activity in senescent cells within the damaged region of human OA cartilage. Autophagy activation may lead to different outcomes depending on the timing, duration, or type of its activation. Thus, our study explored the complex relationship between chondrocyte autophagy and OA, as well as the related regulatory molecules and signaling pathways, providing new insights for the future development of safe and effective drugs targeting chondrocyte autophagy to improve OA.
Collapse
Affiliation(s)
- Lan Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian-Jiang Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Huan Zhou
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xing-Wang Zhu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Ping-Heng Zhang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Huang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen-Ting Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao-Feng Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - En-Sheng Chen
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Cui J, Zhang T, Wang F, Feng L, Deng G, Wu T, Yin L, Hu Y. T1/T2 Proportional Magnetic Resonance Nanoprobe Monitoring Tumor Autophagy during Chemotherapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1673. [PMID: 39453009 PMCID: PMC11510095 DOI: 10.3390/nano14201673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Autophagy leads to cellular tolerance of the therapeutic pressure of chemotherapeutic drugs, resulting in treatment resistance. Therefore, the effective monitoring of the autophagy status of tumors in vivo and the regulating of the autophagy level are crucial for improving the efficacy of chemotherapy. In this work, we grafted nitroxide radicals onto the surface of iron oxide nanoparticles (Fe3O4 NPs) using dendrimer polymers, yielding Fe3O4-NO· NPs that are responsive to reactive oxygen species (ROS) and possess enhanced T1 and T2 signal capabilities in a magnetic resonance imaging (MRI) measurement. The ROS in tumor cells generated by autophagy quenches the nitroxide radicals, thereby weakening the T1 signal. In contrast, Fe3O4 NPs are unaffected by intracellular ROS, leading to a stable T2 signal. By comparing the intensity ratio of T1 to T2 in Fe3O4-NO· NPs, we can evaluate the in vivo autophagy status within tumors in real time. It also revealed that Fe3O4-NO· NPs loaded with doxorubicin (Dox) and combining the autophagy inhibitor exhibited high antitumor activity in cells and tumor-bearing mice. This system, which combines real-time monitoring of tumor cell autophagy with the delivery of chemotherapeutic drugs, provides an innovative and effective strategy for tumor treatment with potential clinical application prospects.
Collapse
Affiliation(s)
- Jia Cui
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; (J.C.); (T.Z.); (F.W.); (L.F.)
| | - Taixing Zhang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; (J.C.); (T.Z.); (F.W.); (L.F.)
| | - Fei Wang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; (J.C.); (T.Z.); (F.W.); (L.F.)
| | - Lingzi Feng
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; (J.C.); (T.Z.); (F.W.); (L.F.)
| | - Guangjun Deng
- Tongzhou Renming Hospital, 999 Jianshe Road, Jinsha Town, Tongzhou District, Nantong 226030, China;
| | - Ting Wu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210068, China
| | - Le Yin
- Tongzhou Renming Hospital, 999 Jianshe Road, Jinsha Town, Tongzhou District, Nantong 226030, China;
| | - Yong Hu
- MOE Key Laboratory of High Performance Polymer Materials and Technology, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210023, China; (J.C.); (T.Z.); (F.W.); (L.F.)
- Nanjing University (Suzhou) High-Tech Institute, Renai Road 150, Suzhou Industrial Park, Suzhou 215123, China
| |
Collapse
|
23
|
Tang L, Ding J, Yang K, Zong Z, Wu R, Li H. New insights into the mechanisms and therapeutic strategies of chondrocyte autophagy in osteoarthritis. J Mol Med (Berl) 2024; 102:1229-1244. [PMID: 39145815 DOI: 10.1007/s00109-024-02473-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/22/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024]
Abstract
Osteoarthritis (OA) is a chronic joint disease with an unclear cause characterized by secondary osteophytes and degenerative changes in the articular cartilage. More than 250 million people are expected to be affected by it by 2050, putting a tremendous socioeconomic strain on the entire world. OA cannot currently be treated with any effective medications that change the illness. Over time, chondrocytes undergo gradual metabolic, structural, and functional changes as a result of aging or abuse. The degenerative progression of osteoarthritis is significantly influenced by the imbalance of chondrocyte homeostasis. By continuously recycling and rebuilding macromolecules or organelles, autophagy functions as a crucial regulatory system to maintain homeostasis during an individual's growth and development. This review uses chondrocytes as its starting point and establishes a strong connection between autophagy and osteoarthritis in order to thoroughly examine the mechanisms behind chondrocyte autophagy in osteoarthritis. Biomarkers of chondrocyte autophagy will be identified, and prospective targeted medications and novel treatment approaches for slowing or preventing the course of OA will be developed based on chondrocyte senescence, autophagy, and apoptosis in OA. KEY MESSAGES: Currently, OA has not been treated with any drugs that can effectively cure it. We hope that by exploring specific targets in the course of osteoarthritis, we can promote the progress of treatment strategies. The degenerative progression of osteoarthritis is significantly influenced by the imbalance of chondrocyte balance. Through the continuous recovery and reconstruction of macromolecules or organelles, autophagy is an important regulatory system for maintaining homeostasis during individual growth and development. In this paper, the close relationship between autophagy and osteoarthritis was established with chondrocytes as the starting point, in order to further explore the mechanism of chondrocyte autophagy in osteoarthritis. The development process of osteoarthritis was studied from the perspective of chondrocytes, and the change of autophagy level had a significant impact on osteoarthritis. Chondrocyte autophagy is mainly determined by intracellular mitochondrial autophagy, so we are committed to finding relevant molecules. Through PI3K/AKT- and MAPK-related pathways, the biomarkers of chondrocyte autophagy were identified, and chondrocyte senescence, autophagy, and apoptosis based on osteoarthritis provided a constructive idea for the development of prospective targeted drugs and new therapies to slow down or prevent the progression of osteoarthritis.
Collapse
Affiliation(s)
- Lujia Tang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
- The Third Clinical Medicine School, Nanchang University, Nanchang, China
| | - Jiatong Ding
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kangping Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Hui Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
24
|
José Alcaraz M. Control of articular degeneration by extracellular vesicles from stem/stromal cells as a potential strategy for the treatment of osteoarthritis. Biochem Pharmacol 2024; 228:116226. [PMID: 38663683 DOI: 10.1016/j.bcp.2024.116226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint condition that contributes to years lived with disability. Current therapeutic approaches are limited as there are no disease-modifying interventions able to delay or inhibit the progression of disease. In recent years there has been an increasing interest in the immunomodulatory and regenerative properties of mesenchymal stem/stromal cells (MSCs) to develop new OA therapies. Extracellular vesicles (EVs) mediate many of the biological effects of these cells and may represent an alternative avoiding the limitations of cell-based therapy. There is also a growing interest in EV modifications to enhance their efficacy and applications. Recent preclinical studies have provided strong evidence supporting the potential of MSC EVs for the development of OA treatments. Thus, MSC EVs may regulate chondrocyte functions to avoid cartilage destruction, inhibit abnormal subchondral bone metabolism and synovial tissue alterations, and control pain behavior. EV actions may be mediated by the transfer of their cargo to target cells, with an important role for proteins and non-coding RNAs modulating signaling pathways relevant for OA progression. Nevertheless, additional investigations are needed concerning EV optimization, and standardization of preparation procedures. More research is also required for a better knowledge of possible effects on different OA phenotypes, pharmacokinetics, mechanism of action, long-term effects and safety profile. Furthermore, MSC EVs have a high potential as vehicles for drug delivery or as adjuvant therapy to potentiate or complement the effects of other approaches.
Collapse
Affiliation(s)
- María José Alcaraz
- Department of Pharmacology, University of Valencia, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| |
Collapse
|
25
|
Zhang Y, Zhou Y. Advances in targeted therapies for age-related osteoarthritis: A comprehensive review of current research. Biomed Pharmacother 2024; 179:117314. [PMID: 39167845 DOI: 10.1016/j.biopha.2024.117314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that disproportionately impacts the elderly population on a global scale. As aging is a significant risk factor for OA, there is a growing urgency to develop specific therapies that target the underlying mechanisms of aging associated with this condition. This summary seeks to offer a thorough introduction of ongoing research efforts aimed at developing therapies to combat senescence in the context of OA. Cellular senescence plays a pivotal role in both the deterioration of cartilage integrity and the perpetuation of chronic inflammation and tissue remodeling. Consequently, targeting SnCs has emerged as a promising therapeutic approach to alleviate symptoms and hinder the progression of OA. This review examines a range of approaches, including senolytic drugs targeting SnCs, senomorphics that modulate the senescence-associated secretory phenotype (SASP), and interventions that enhance immune system clearance of SnCs. Novel methodologies, such as utilizing novel materials for exosome delivery and administering anti-aging medications with precision, offer promising avenues for the precise treatment of OA. Accumulating evidence underscores the potential of targeting senescence in OA management, potentially facilitating the development of more effective and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Yantao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuan 430060, China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuan 430060, China.
| |
Collapse
|
26
|
Xu T, Zhang K, Hu Y, Yang R, Tang J, Fu W. Comparison of the Therapeutic Efficacy and Autophagy-Mediated Mechanisms of Action of Urine-Derived and Adipose-Derived Stem Cells in Osteoarthritis. Am J Sports Med 2024; 52:3130-3146. [PMID: 39311500 DOI: 10.1177/03635465241277176] [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] [Indexed: 10/22/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent and disabling disease that affects a significant proportion of the global population. Urine-derived stem cells (USCs) have shown great prospects in the treatment of OA, but there is no study that has compared them with traditional stem cells. PURPOSE This study aimed to compare the therapeutic efficacy and mechanisms of USCs and adipose-derived stem cells (ADSCs) for OA treatment. STUDY DESIGN Controlled laboratory study. METHODS We compared the biological properties of USCs and ADSCs using CCK-8, colony formation, EdU, adhesion, and apoptosis assays. We evaluated the protective effects of USCs and ADSCs on IL-1β-treated OA chondrocytes by chemical staining, immunofluorescence, and Western blotting. We assessed the effects of USCs and ADSCs on chondrocyte autophagy by transmission electron microscopy, immunofluorescence, and Western blotting. We also compared the therapeutic efficacy of intra-articular injections of USCs and ADSCs by gross, histological, micro-computed tomography, and immunohistochemical analyses in an OA rat model induced by anterior cruciate ligament transection. RESULTS USCs showed higher proliferation, colony formation, DNA synthesis, adhesion, and anti-apoptotic abilities than ADSCs. Both USCs and ADSCs increased the expression of cartilage-specific proteins and decreased the expression of matrix degradation-related proteins and inflammatory factors in OA chondrocytes. USCs had a greater advantage in suppressing MMP-13 and inflammatory factors than ADSCs. Both USCs and ADSCs enhanced autophagy in OA chondrocytes, with USCs being more effective than ADSCs. The autophagy inhibitor 3-MA reduced the enhanced autophagy and protective effects of USCs and ADSCs on OA chondrocytes. CONCLUSION To our knowledge, this is the first study to explore the efficacy of USCs in the treatment of knee OA and to compare them with ADSCs. Considering the superior properties of USCs in terms of noninvasive acquisition, a high cost-benefit ratio, and low ethical concerns, our study suggests that they may be a more promising therapeutic option than ADSCs for OA treatment under rigorous regulatory pathways. CLINICAL RELEVANCE USCs may be a superior cell source for stem cells to treat knee OA, and this study strengthens the evidence for the application of USCs.
Collapse
Affiliation(s)
- Tianhao Xu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaibo Zhang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunan Hu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Runze Yang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiexi Tang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weili Fu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
27
|
Wu J, Huang S, Yu Y, Lian Q, Liu Y, Dai W, Liu Q, Pan Y, Liu GA, Li K, Liu C, Li G. Human adipose and synovial-derived MSCs synergistically attenuate osteoarthritis by promoting chondrocyte autophagy through FoxO1 signaling. Stem Cell Res Ther 2024; 15:261. [PMID: 39148121 PMCID: PMC11328463 DOI: 10.1186/s13287-024-03870-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Human adipose-derived stem cells (ADSCs) exert a strong anti-inflammatory effect, and synovium-derived stem cells (SDSCs) have high chondrogenic potential. Thus, this study aims to investigate whether a combination of human ADSCs and SDSCs will have a synergistic effect that will increase the chondrogenic potential of osteoarthritis (OA) chondrocytes in vitro and attenuate the cartilage degeneration of early and advanced OA in vitro. METHODS ADSCs, SDSCs, and chondrocytes were isolated from OA patients who underwent total knee arthroplasty. The ADSCs-SDSCs mixed cell ratios were 1:0 (ADSCs only), 8:2, 5:5 (5A5S), 2:8, and 0:1 (SDSCs only). The chondrogenic potential of the OA chondrocytes was evaluated in vitro with a transwell assay or pellet culture with various mixed cell groups. The mixed cell group with the highest chondrogenic potential was then selected and injected into the knee joints of nude rats of early and advanced OA stages in vivo. The animals were then evaluated 12 and 20 weeks after surgery through gait analysis, von frey test, microcomputed tomography, MRI, and immunohistochemical and histological analyses. Finally, the mechanisms underlying these findings were investigated through the RNA sequencing of tissue samples in vivo and Western blot of the OA chondrocyte autophagy pathway. RESULTS Among the MSCs treatment groups, 5A5S had the greatest synergistic effect that increased the chondrogenic potential of OA chondrocytes in vitro and inhibited early and advanced OA in vivo. The 5A5S group significantly reduced cartilage degeneration, synovial inflammation, pain sensation, and nerve invasion in subchondral nude rat OA, outperforming both single-cell treatments. The underlying mechanism was the activation of chondrocyte autophagy via the FoxO1 signaling pathway. CONCLUSION A combination of human ADSCs and SDSCs demonstrated higher potential than a single type of stem cell, demonstrating potential as a novel treatment for OA.
Collapse
Affiliation(s)
- Jianqun Wu
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China
| | - Songqiang Huang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, Hunan, China
| | - Yangyi Yu
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China
| | - Qiang Lian
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Wenfeng Dai
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China
| | - Qisong Liu
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China
| | - Yonghao Pan
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Gui-Ang Liu
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China
| | - Kai Li
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Guangheng Li
- Division of Adult Joint Reconstruction and Sports Medicine, Department of Orthopedic Surgery, The First Affiliated Hospital (Shenzhen People's Hospital),, School of Medicine, Southern University of Science and Technology, 1017 Dongmen North Road, Luohu District, Shenzhen, 518055, China.
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen People's Hospital, Guangdong, China.
| |
Collapse
|
28
|
Lawrence M, Goyal A, Pathak S, Ganguly P. Cellular Senescence and Inflammaging in the Bone: Pathways, Genetics, Anti-Aging Strategies and Interventions. Int J Mol Sci 2024; 25:7411. [PMID: 39000517 PMCID: PMC11242738 DOI: 10.3390/ijms25137411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Advancing age is associated with several age-related diseases (ARDs), with musculoskeletal conditions impacting millions of elderly people worldwide. With orthopedic conditions contributing towards considerable number of patients, a deeper understanding of bone aging is the need of the hour. One of the underlying factors of bone aging is cellular senescence and its associated senescence associated secretory phenotype (SASP). SASP comprises of pro-inflammatory markers, cytokines and chemokines that arrest cell growth and development. The accumulation of SASP over several years leads to chronic low-grade inflammation with advancing age, also known as inflammaging. The pathways and molecular mechanisms focused on bone senescence and inflammaging are currently limited but are increasingly being explored. Most of the genes, pathways and mechanisms involved in senescence and inflammaging coincide with those associated with cancer and other ARDs like osteoarthritis (OA). Thus, exploring these pathways using techniques like sequencing, identifying these factors and combatting them with the most suitable approach are crucial for healthy aging and the early detection of ARDs. Several approaches can be used to aid regeneration and reduce senescence in the bone. These may be pharmacological, non-pharmacological and lifestyle interventions. With increasing evidence towards the intricate relationship between aging, senescence, inflammation and ARDs, these approaches may also be used as anti-aging strategies for the aging bone marrow (BM).
Collapse
Affiliation(s)
- Merin Lawrence
- School of Biological and Chemical Sciences, University of Galway, H91W2TY Galway, Ireland
| | - Abhishek Goyal
- RAS Life Science Solutions, Stresemannallee 61, 60596 Frankfurt, Germany
| | - Shelly Pathak
- Observational and Pragmatic Research Institute, 5 Coles Lane, Oakington, Cambridge CB24 3BA, UK
| | - Payal Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK
| |
Collapse
|
29
|
Cai H, Zheng Y, Chen Y, Lu Q, Hong W, Guo Q, Zheng S. Miao medicine Gu Yan Xiao tincture inhibits mTOR to stimulate chondrocyte autophagy in a rabbit model of osteoarthritis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118095. [PMID: 38548121 DOI: 10.1016/j.jep.2024.118095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/10/2024] [Accepted: 03/21/2024] [Indexed: 04/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Gu Yan Xiao tincture, a blend of traditional Chinese herbs, is traditionally used for osteoarthritis and related pain. This study investigated its mechanism of action in order to rationalize and validate its therapeutic use. AIM OF THE STUDY This study analyzed, in a rabbit model of knee osteoarthritis, whether and how Gu Yan Xiao tincture exerts therapeutic benefits by modulating chondrocyte autophagy. MATERIALS AND METHODS The active constituents within the GYX tincture were identified using liquid chromatography-mass spectrometry. The rabbit model was established by injecting animals with type II collagenase intra-articularly, and the effects of topically applied tincture were examined on osteoarthritis lesions of the knee using histopathology, micro-computed tomography and x-ray imaging. Effects of the tincture were also evaluated on levels of inflammatory cytokines, matrix metalloproteases, and autophagy in chondrocytes. As a positive control, animals were treated with sodium diclofenac. RESULTS The tincture mitigated the reduction in joint space, hyperplasia of the synovium and matrix metalloproteases in serum that occurred after injection of type II collagenase in rabbits. These therapeutic effects were associated with inhibition of mTOR and activation of autophagy in articular chondrocytes. Inhibiting mTOR with rapamycin potentiated the therapeutic effects of the tincture, while inhibiting autophagy with 3-methyladenine antagonized them. CONCLUSIONS Gu Yan Xiao tincture mitigates tissue injury in a rabbit model of osteoarthritis, at least in part by inhibiting mTOR and thereby promoting autophagy in chondrocytes. These results rationalize the use of the tincture not only against osteoarthritis but also potentially other diseases involving inhibition of autophagy in bones and joints.
Collapse
Affiliation(s)
- He Cai
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Yuhao Zheng
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Yinying Chen
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Qing Lu
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Wu Hong
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Qiucheng Guo
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Shuguang Zheng
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China; The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| |
Collapse
|
30
|
Liu Y, Wang Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanotherapeutics targeting autophagy regulation for improved cancer therapy. Acta Pharm Sin B 2024; 14:2447-2474. [PMID: 38828133 PMCID: PMC11143539 DOI: 10.1016/j.apsb.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/29/2024] [Indexed: 06/05/2024] Open
Abstract
The clinical efficacy of current cancer therapies falls short, and there is a pressing demand to integrate new targets with conventional therapies. Autophagy, a highly conserved self-degradation process, has received considerable attention as an emerging therapeutic target for cancer. With the rapid development of nanomedicine, nanomaterials have been widely utilized in cancer therapy due to their unrivaled delivery performance. Hence, considering the potential benefits of integrating autophagy and nanotechnology in cancer therapy, we outline the latest advances in autophagy-based nanotherapeutics. Based on a brief background related to autophagy and nanotherapeutics and their impact on tumor progression, the feasibility of autophagy-based nanotherapeutics for cancer treatment is demonstrated. Further, emerging nanotherapeutics developed to modulate autophagy are reviewed from the perspective of cell signaling pathways, including modulation of the mammalian target of rapamycin (mTOR) pathway, autophagy-related (ATG) and its complex expression, reactive oxygen species (ROS) and mitophagy, interference with autophagosome-lysosome fusion, and inhibition of hypoxia-mediated autophagy. In addition, combination therapies in which nano-autophagy modulation is combined with chemotherapy, phototherapy, and immunotherapy are also described. Finally, the prospects and challenges of autophagy-based nanotherapeutics for efficient cancer treatment are envisioned.
Collapse
Affiliation(s)
- Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, China
| |
Collapse
|
31
|
Kong P, Ahmad RE, Zulkifli A, Krishnan S, Nam HY, Kamarul T. The role of autophagy in mitigating osteoarthritis progression via regulation of chondrocyte apoptosis: A review. Joint Bone Spine 2024; 91:105642. [PMID: 37739213 DOI: 10.1016/j.jbspin.2023.105642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/22/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease with an immense socioeconomic burden; however, no treatment has achieved complete success in effectively halting or reversing cartilage degradation, which is the central pathophysiological feature of OA. Chondrocytes loss or dysfunction is a significant contributing factor to the progressive cartilage deterioration as these sole resident cells have a crucial role to produce extracellular matrix proteins, thus maintaining cartilage structure and homeostasis. It has been previously suggested that death of chondrocytes occurring through apoptosis substantially contributes to cartilage degeneration. Although the occurrence of apoptosis in osteoarthritic cartilage and its correlation with cartilage degradation is evident, the causes of chondrocyte apoptosis leading to matrix loss are still not well-understood. Autophagy, an intracellular degradative mechanism that eliminates dysfunctional cytoplasmic components to aid cell survival in unfavourable conditions, is a potential therapeutic target to inhibit chondrocyte apoptosis and reduce OA severity. Despite accumulating evidence indicating significant cytoprotective effects of autophagy against chondrocyte apoptosis, the mechanistic link between autophagy and apoptosis in chondrocytes remains to be further explored. In this review, we summarize the relevant mechanistic events that perpetuate chondrocyte apoptosis and highlight the prominent role of autophagy in modulating these events to mitigate OA progression.
Collapse
Affiliation(s)
- Peggy Kong
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Raja Elina Ahmad
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia.
| | - Amirah Zulkifli
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Shaliny Krishnan
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Hui Yin Nam
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia; Nanotechnology and Catalysis Research Centre (NANOCAT), Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia; Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas Pulau Pinang, Malaysia
| |
Collapse
|
32
|
Ma T, Xu G, Gao T, Zhao G, Huang G, Shi J, Chen J, Song J, Xia J, Ma X. Engineered Exosomes with ATF5-Modified mRNA Loaded in Injectable Thermogels Alleviate Osteoarthritis by Targeting the Mitochondrial Unfolded Protein Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21383-21399. [PMID: 38626424 DOI: 10.1021/acsami.3c17209] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Osteoarthritis (OA) progression is highly associated with chondrocyte mitochondrial dysfunction and disorders of catabolism and anabolism of the extracellular matrix (ECM) in the articular cartilage. The mitochondrial unfolded protein response (UPRmt), which is an integral component of the mitochondrial quality control (MQC) system, is essential for maintaining chondrocyte homeostasis. We successfully validated the pivotal role of activating transcription factor 5 (ATF5) in upregulating the UPRmt, mitigating IL-1β-induced inflammation and mitochondrial dysfunction, and promoting balanced metabolism in articular cartilage ECM, proving its potential as a promising therapeutic target for OA. Modified mRNAs (modRNAs) have emerged as novel and efficient gene delivery vectors for nucleic acid therapeutic approaches. In this study, we combined Atf5-modRNA (modAtf5) with engineered exosomes derived from bone mesenchymal stem cells (ExmodAtf5) to exert cytoprotective effects on chondrocytes in articular cartilage via Atf5. However, the rapid localized metabolization of ExmodAtf5 limits its application. PLGA-PEG-PLGA (Gel), an injectable thermosensitive hydrogel, was used as a carrier of ExmodAtf5 (Gel@ExmodAtf5) to achieve a sustained release of ExmodAtf5. In vitro and in vivo, the use of Gel@ExmodAtf5 was shown to be a highly effective strategy for OA treatment. The in vivo therapeutic effect of Gel@ExmodAtf5 was evidenced by the preservation of the intact cartilage surface, low OARSI scores, fewer osteophytes, and mild subchondral bone sclerosis and cystic degeneration. Consequently, the combination of ExmodAtf5 and PLGA-PEG-PLGA could significantly enhance the therapeutic efficacy and prolong the exosome release. In addition, the mitochondrial protease ClpP enhanced chondrocyte autophagy by modulating the mTOR/Ulk1 pathway. As a result of our research, Gel@ExmodAtf5 can be considered to be effective at alleviating the progression of OA.
Collapse
Affiliation(s)
- Tiancong Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guangyu Xu
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Tian Gao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Guanglei Zhao
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Gangyong Huang
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jingsheng Shi
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jie Chen
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jian Song
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Jun Xia
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| | - Xiaosheng Ma
- Department of Orthopaedic Surgery, Huashan Hospital Fudan University, 12th Wulumuqi Middle Road, Jing'an District, Shanghai 200040, China
- Fudan University, 220th Handan Road, Yang'pu District, Shanghai 200082, China
| |
Collapse
|
33
|
Chen B, Sun Y, Xu G, Jiang J, Zhang W, Wu C, Xue P, Cui Z. Role of crosstalk between synovial cells and chondrocytes in osteoarthritis (Review). Exp Ther Med 2024; 27:201. [PMID: 38590580 PMCID: PMC11000048 DOI: 10.3892/etm.2024.12490] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 04/10/2024] Open
Abstract
Osteoarthritis (OA) is a low-grade, nonspecific inflammatory disease that affects the entire joint. This condition is characterized by synovitis, cartilage erosion, subchondral bone defects, and subpatellar fat pad damage. There is mounting evidence demonstrating the significance of crosstalk between synovitis and cartilage destruction in the development of OA. To comprehensively explore the phenotypic alterations of synovitis and cartilage destruction, it is important to elucidate the crosstalk mechanisms between chondrocytes and synovial cells. Furthermore, the updated iteration of single-cell sequencing technology reveals the interaction between chondrocyte and synovial cells. In the present review, the histological and pathological alterations between cartilage and synovium during OA progression are described, and the mode of interaction and molecular mechanisms between synovial cells and chondrocytes in OA, both of which affect the OA process mainly by altering the inflammatory environment and cellular state, are elucidated. Finally, the current OA therapeutic approaches are summarized and emerging therapeutic targets are reviewed in an attempt to provide potential insights into OA treatment.
Collapse
Affiliation(s)
- Baisen Chen
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yuyu Sun
- Department of Orthopedics, Nantong Third People's Hospital, Nantong, Jiangsu 226003, P.R. China
| | - Guanhua Xu
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jiawei Jiang
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wenhao Zhang
- Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chunshuai Wu
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Pengfei Xue
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhiming Cui
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
34
|
Sulcanese L, Prencipe G, Canciello A, Cerveró-Varona A, Perugini M, Mauro A, Russo V, Barboni B. Stem-Cell-Driven Chondrogenesis: Perspectives on Amnion-Derived Cells. Cells 2024; 13:744. [PMID: 38727280 PMCID: PMC11083072 DOI: 10.3390/cells13090744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Regenerative medicine harnesses stem cells' capacity to restore damaged tissues and organs. In vitro methods employing specific bioactive molecules, such as growth factors, bio-inductive scaffolds, 3D cultures, co-cultures, and mechanical stimuli, steer stem cells toward the desired differentiation pathways, mimicking their natural development. Chondrogenesis presents a challenge for regenerative medicine. This intricate process involves precise modulation of chondro-related transcription factors and pathways, critical for generating cartilage. Cartilage damage disrupts this process, impeding proper tissue healing due to its unique mechanical and anatomical characteristics. Consequently, the resultant tissue often forms fibrocartilage, which lacks adequate mechanical properties, posing a significant hurdle for effective regeneration. This review comprehensively explores studies showcasing the potential of amniotic mesenchymal stem cells (AMSCs) and amniotic epithelial cells (AECs) in chondrogenic differentiation. These cells exhibit innate characteristics that position them as promising candidates for regenerative medicine. Their capacity to differentiate toward chondrocytes offers a pathway for developing effective regenerative protocols. Understanding and leveraging the innate properties of AMSCs and AECs hold promise in addressing the challenges associated with cartilage repair, potentially offering superior outcomes in tissue regeneration.
Collapse
Affiliation(s)
- Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Monia Perugini
- Department of Bioscience and Technology for Food, Agriculture, and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agri-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (G.P.); (A.C.); (A.C.-V.); (A.M.); (V.R.); (B.B.)
| |
Collapse
|
35
|
Wei Q, Yu Z, Yang P, Chen X. Baicalin Maintains Articular Cartilage Homeostasis and Alleviates Osteoarthritis by Activating FOXO1. J Med Food 2024; 27:301-311. [PMID: 38377551 DOI: 10.1089/jmf.2023.k.0206] [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: 02/22/2024] Open
Abstract
Baicalin has been acknowledged for its anti-inflammatory properties. However, its potential impact on osteoarthritis (OA) has not yet been explored. Therefore, our study aimed to examine the effects of Baicalin on OA, both in laboratory and animal models. To evaluate its efficacy, human chondrocytes affected by OA were treated with interleukin-1β and/or Baicalin. The effects were then assessed through viability tests using the cell counting kit-8 (CCK-8) method and flow cytometry. In addition, we analyzed the expressions of various factors such as FOXO1, autophagy, apoptosis, and cartilage synthesis and breakdown to corroborate the effects of Baicalin. We also assessed the severity of OA through analysis of tissue samples. Our findings demonstrate that Baicalin effectively suppresses inflammatory cytokines and MMP-13 levels caused by collagenase-induced osteoarthritis, while simultaneously preserving the levels of Aggrecan and Col2. Furthermore, Baicalin has been shown to enhance autophagy. Through the use of FOXO1 inhibitors, lentivirus-mediated knockdown, and chromatin immunoprecipitation, we verified that Baicalin exerts its protective effects by activating FOXO1, which binds to the Beclin-1 promoter, thereby promoting autophagy. In conclusion, our results show that Baicalin has potential as a therapeutic agent for treating OA (Clinical Trial Registration number: 2023-61).
Collapse
Affiliation(s)
- Qiang Wei
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Zhaoping Yu
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Peng Yang
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Xiaohu Chen
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| |
Collapse
|
36
|
Liu L, Wang J, Liu L, Shi W, Gao H, Liu L. WITHDRAWN: The dysregulated autophagy in osteoarthritis: Revisiting molecular profile. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024:S0079-6107(24)00034-8. [PMID: 38531488 DOI: 10.1016/j.pbiomolbio.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/21/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.
Collapse
Affiliation(s)
- Liang Liu
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Jie Wang
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Lu Liu
- Department of Internal Medicine, Tianbao Central Health Hospital, Xintai City, Shandong Province, Shandong, Xintai, 271200, China
| | - Wenling Shi
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Huajie Gao
- Operating Room of Qingdao University Affiliated Hospital, Qingdao, Pingdu, 266000, China
| | - Lun Liu
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| |
Collapse
|
37
|
Lu J, Bian J, Wang Y, Zhao Y, Zhao X, Wang G, Yang J. Oxymatrine protects articular chondrocytes from IL-1β-induced damage through autophagy activation via AKT/mTOR signaling pathway inhibition. J Orthop Surg Res 2024; 19:178. [PMID: 38468339 PMCID: PMC10926585 DOI: 10.1186/s13018-024-04667-2] [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: 08/21/2023] [Accepted: 03/06/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common degenerative joint disease characterized by persistent articular cartilage degeneration and synovitis. Oxymatrine (OMT) is a quinzolazine alkaloid extracted from the traditional Chinese medicine, matrine, and possesses anti-inflammatory properties that may help regulate the pathogenesis of OA; however, its mechanism has not been elucidated. This study aimed to investigate the effects of OMT on interleukin-1β (IL-1β)-induced damage and the potential mechanisms of action. METHODS Chondrocytes were isolated from Sprague-Dawley rats. Toluidine blue and Collagen II immunofluorescence staining were used to determine the purity of the chondrocytes. Thereafter, the chondrocytes were subjected to IL-1β stimulation, both in the presence and absence of OMT, or the autophagy inhibitor 3-methyladenine (3-MA). Cell viability was assessed using the MTT assay and SYTOX Green staining. Additionally, flow cytometry was used to determine cell apoptosis rate and reactive oxygen species (ROS) levels. The protein levels of AKT, mTOR, LC3, P62, matrix metalloproteinase-13, and collagen II were quantitatively analyzed using western blotting. Immunofluorescence was used to assess LC3 expression. RESULTS OMT alleviated IL-1β-induced damage in chondrocytes, by increasing the survival rate, reducing the apoptosis rates of chondrocytes, and preventing the degradation of the cartilage matrix. In addition, OMT decreased the ROS levels and inhibited the AKT/mTOR signaling pathway while promoting autophagy in IL-1β treated chondrocytes. However, the effectiveness of OMT in improving chondrocyte viability under IL-1β treatment was limited when autophagy was inhibited by 3-MA. CONCLUSIONS OMT decreases oxidative stress and inhibits the AKT/mTOR signaling pathway to enhance autophagy, thus inhibiting IL-1β-induced damage. Therefore, OMT may be a novel and effective therapeutic agent for the clinical treatment of OA.
Collapse
Affiliation(s)
- Jinying Lu
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Jiang Bian
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Yutong Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Yan Zhao
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Xinmin Zhao
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Gao Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China
| | - Jing Yang
- Provincial Key Laboratory of Cardiovascular and Cerebrovascular Drug Basic Research, Jinzhou Medical University, No.40, Section 3 Songpo Road, Linghe District, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
38
|
Tang K, Sun L, Chen L, Feng X, Wu J, Guo H, Zheng Y. Bioinformatics Analysis and Experimental Validation of Mitochondrial Autophagy Genes in Knee Osteoarthritis. Int J Gen Med 2024; 17:639-650. [PMID: 38414629 PMCID: PMC10898481 DOI: 10.2147/ijgm.s444847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Background Mitochondrial autophagy is closely related to the pathogenesis of osteoarthritis, In order to explore the role of mitochondrial autophagy related genes in knee osteoarthritis (KOA) and its molecular mechanism. Methods KOA-related transcriptome data were extracted from the Gene Expression Omnibus (GEO) database. Differentially expressed mitochondrial autophagy gene (DEMGs) were screened in patients with KOA by differential expression analysis. The STRING website was used to construct a protein-protein interaction (PPI) network among DEMGs. Molecular complex detection (MCODE) method in Cytoscape software was performed to identify hub DEMGs. Support vector machine recursive feature elimination (SVM-RFE) method was used to construct the hub DEMG diagnosis model. Genes with diagnostic value were identified as biomarkers by plotting receiver operating characteristic (ROC) curves and Expression validation. CIBERSORT algorithm was used to calculate the proportion of 22 immune cells in each sample in the GSE114007 dataset. Finally, biomarker expression was verified by qPCR. Results A total of 15 DEMGs were obtained and enrichment analyses showed that these DEMG strains were mainly enriched in the mitophagy-animal, shigellosis, autophagy-animal and FoxO signal pathways. The PPI network unveiled 13 DEMGs with interactions. In addition, 8 hub DEMGs (ULK1, CALCOCO2, MAP1LC3B, BNIP3L, GABARAPL1, BNIP3, FKBP8 and FOXO3) were obtained for KOA. And 5 model DEMGs (BNIP3L, BNIP3, MAP1LC3B, ULK1 and FOXO3) were screened. The ROC curves revealed that BNIP3 and FOXO3 has strong diagnostic value in these models of DEMG. Immune-infiltration and correlation analysis showed that BNIP3 and FOXO3 were significantly correlated with three different immune cells, including primary B cells, M0 macrophage and M2 macrophage. The cartilage tissue samples qPCR verification results show that FOXO3 and BNIP3 were all down-regulated in KOA (p < 0.01), and the validation results are consistent with the above analysis. Conclusion BNIP3 and FOXO3 have been identified as biomarkers for the diagnosis of KOA, which might supply a new insight for the pathogenesis and treatment of KOA.
Collapse
Affiliation(s)
- Kuihan Tang
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People’s Republic of China
| | - Li Sun
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Long Chen
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Xiaobo Feng
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People’s Republic of China
| | - Jiarui Wu
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Hao Guo
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Yong Zheng
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People’s Republic of China
| |
Collapse
|
39
|
Zhou X, Li WK, Zhuang C, Zhou XC, Zhao XF, Pan Y, Guo WX, Yang YW, Sheng CZ, Xie ZF, Yu JS, Chen YX, Wang LK, Ma TY, Zhu KX, Xiang KM, Zhuang RJ. Lei's formula attenuates osteoarthritis mediated by suppression of chondrocyte senescence via the mTOR axis: in vitro and in vivo experiments. Aging (Albany NY) 2024; 16:4250-4269. [PMID: 38407978 PMCID: PMC10968702 DOI: 10.18632/aging.205582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
Lei's formula (LSF), a traditional Chinese herbal remedy, is recognized for its remarkable clinical effectiveness in treating osteoarthritis (OA). Despite its therapeutic potential, the exact molecular mechanisms underlying LSF's action in OA have remained enigmatic. Existing research has shed light on the role of the mTOR signaling pathway in promoting chondrocyte senescence, a central factor in OA-related cartilage degeneration. Consequently, targeting mTOR to mitigate chondrocyte senescence presents a promising avenue for OA treatment. The primary objective of this study is to establish LSF's chondroprotective potential and confirm its anti-osteoarthritic efficacy through mTOR inhibition. In vivo assessments using an OA mouse model reveal substantial articular cartilage degeneration. However, LSF serves as an effective guardian of articular cartilage, evidenced by reduced subchondral osteosclerosis, increased cartilage thickness, improved surface smoothness, decreased OARSI scores, elevated expression of cartilage anabolic markers (Col2 and Aggrecan), reduced expression of catabolic markers (Adamts5 and MMP13), increased expression of the chondrocyte hypertrophy marker (Col10), and decreased expression of chondrocyte senescence markers (P16 and P21). In vitro findings demonstrate that LSF shields chondrocytes from H2O2-induced apoptosis, inhibits senescence, enhances chondrocyte differentiation, promotes the synthesis of type II collagen and proteoglycans, and reduces cartilage degradation. Mechanistically, LSF suppresses chondrocyte senescence through the mTOR axis, orchestrating the equilibrium between chondrocyte anabolism and catabolism, ultimately leading to reduced apoptosis and decelerated OA cartilage degradation. LSF holds significant promise as a therapeutic approach for OA treatment, offering new insights into potential treatments for this prevalent age-related condition.
Collapse
Affiliation(s)
- Xing Zhou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wen-Kai Li
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chen Zhuang
- Alberta Institute, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing-Chen Zhou
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xue-Fei Zhao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yu Pan
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wen-Xuan Guo
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yi-Wen Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Cen-Zhuo Sheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhe-Fei Xie
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jin-Sheng Yu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yi-Xuan Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Li-Kang Wang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tian-You Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kang-Xiang Zhu
- Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, China
- Quzhou TCM Hospital at the Junction of Four Provinces Affiliated to Zhejiang Chinese Medical University, Quzhou, Zhejiang, China
| | - Ke-Meng Xiang
- Taizhou Traditional Chinese Medicine Hospital, Taizhou, Zhejiang, China
| | - Ru-Jie Zhuang
- Department of Orthopaedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, China
- Quzhou TCM Hospital at the Junction of Four Provinces Affiliated to Zhejiang Chinese Medical University, Quzhou, Zhejiang, China
| |
Collapse
|
40
|
Xu W, Gu S, Zhang G, Wang R, Lv S, Yan J, Qin Y. APOD acts on fibroblast-like synoviocyte and chondrocyte to alleviate the process of osteoarthritis in vitro. J Orthop Res 2024; 42:296-305. [PMID: 37728985 DOI: 10.1002/jor.25690] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/03/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
The pathogenesis of osteoarthritis (OA) is still unclear, leading to the lack of targeted treatment. We aimed to probe into the effect of apolipoprotein D (APOD), the key gene from our previous study through bioinformatics analysis, on fibroblast-like synoviocyte (FLS) and chondrocytes in vitro to confirm its potential roles on the delay of OA progression. Primary FLS and chondrocytes were extracted from synovium and cartilage of OA patients and stimulated with interleukin 1β (IL-1β) in vitro. After APOD intervention, viability and proliferation of FLS and chondrocytes were detected. Subsequently, the inflammatory factors of the two cells were detected by quantitative reverse-transcription polymerase chain reaction, enzyme-linked immunosorbent assay, and western blot, and the apoptosis and autophagy-related substances were determined at the same time. Finally, the oxidation level in FLS and chondrocytes were detected. APOD reversed the change of gene expression stimulated by IL-1β in FLS and chondrocytes. APOD alleviated the proliferation of FLS while promoted proliferation of chondrocytes, and reduced the expression of inflammatory factors. Moreover, APOD promoted apoptosis of FLS and autography of chondrocytes, while reduced apoptosis of chondrocytes. Finally, decrease level of reactive oxygen species (ROS) in both cells were observed after APOD intervention, as well as the increased expression of antioxidant-related genes. APOD had effects on the proliferation of FLS and chondrocytes through apoptosis and autography as well as the reduction of oxidative stress, delaying the progress of OA.
Collapse
Affiliation(s)
- Wenbo Xu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shoubin Gu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ren Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Songcen Lv
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Qin
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
41
|
Xiong Z, Peng G, Deng J, Liu M, Ning X, Zhuang Y, Yang H, Sun H. Therapeutic targets and potential delivery systems of melatonin in osteoarthritis. Front Immunol 2024; 15:1331934. [PMID: 38327517 PMCID: PMC10847247 DOI: 10.3389/fimmu.2024.1331934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/04/2024] [Indexed: 02/09/2024] Open
Abstract
Osteoarthritis (OA) is a highly prevalent age-related musculoskeletal disorder that typically results in chronic pain and disability. OA is a multifactorial disease, with increased oxidative stress, dysregulated inflammatory response, and impaired matrix metabolism contributing to its onset and progression. The neurohormone melatonin, primarily synthesized by the pineal gland, has emerged as a promising therapeutic agent for OA due to its potential to alleviate inflammation, oxidative stress, and chondrocyte death with minimal adverse effects. The present review provides a comprehensive summary of the current understanding regarding melatonin as a promising pharmaceutical agent for the treatment of OA, along with an exploration of various delivery systems that can be utilized for melatonin administration. These findings may provide novel therapeutic strategies and targets for inhibiting the advancement of OA.
Collapse
Affiliation(s)
- Zhilin Xiong
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guoxuan Peng
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jin Deng
- Department of Emergence Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Miao Liu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xu Ning
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong Zhuang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hua Yang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hong Sun
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Emergence Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
42
|
Yuan D, Shen H, Bai L, Li M, Ye Q. Identification of Key Ubiquitination-Related Genes and Their Association with Immune Infiltration in Osteoarthritis Based on the mRNA-miRNA Network. Crit Rev Immunol 2024; 44:91-102. [PMID: 38505924 DOI: 10.1615/critrevimmunol.2024051440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease that is closely associated with functions of ubiquitination and immune cells, yet the mechanism remains ambiguous. This study aimed to find core ubiquitination-related genes and their correlative immune infiltration in OA using weighted gene co-expression network analysis (WGCNA). The ubiquitination-related genes, datasets GSE55235 and GSE143514 were obtained from open databases. WGCNA got used to investigate key co-expressed genes. Then, we screened differentially expressed miRNAs by "limma" package in R, and constructed mRNA-miRNA network. We conducted function enrichment analysis on the identified genes. CIBERSORT was then utilized to analyze the relevance between immune infiltration and genes. Lastly, RT-qPCR was further used to verify the prediction of bioinformatics. A sum of 144 ubiquitination-related genes in OA were acquired. Enrichment analysis indicated that obtained genes obviously involved in mTOR pathway to regulate the OA development. GRB2 and SEH1L and L-arginine synergistically regulate the mTOR signaling pathway in OA. Moreover, GRB2 and SEH1L were remarkably bound up with immune cell infiltration. Additionally, GRB2 expression was upregulated and SEH1L level was downregulated in the OA development by RT-qPCR experiment. The present study identified GRB2 and SEH1L as key ubiquitination-related genes which were involved in immune infiltration in OA patients, thereby providing new drug targets for OA.
Collapse
Affiliation(s)
- Dalu Yuan
- Department of Rehabilitation Treatment Center, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, Zhejiang, China
| | - Hailiang Shen
- Department of Orthopedics, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, Zhejiang, China
| | - Lina Bai
- Department of Rehabilitation Treatment Center, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, Zhejiang, China
| | - Menglin Li
- Department of Rehabilitation Treatment Center, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), Shaoxing, Zhejiang, China
| | - Qiujie Ye
- Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital)
| |
Collapse
|
43
|
Cheng C, Wu Y, Huang Y, Xue Q, Wang Y, Liao F, Wang X, Miao C. Epigenetic modification and exosome effects on autophagy in osteoarthritis. Biochem Pharmacol 2023; 218:115930. [PMID: 37979704 DOI: 10.1016/j.bcp.2023.115930] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease that leads to joint pain and stiffness and is one of the leading causes of disability and pain worldwide. Autophagy is a highly conserved self-degradation process, and its abnormal function is closely related to human diseases, including OA. Abnormal autophagy regulates cell aging, matrix metalloproteinase metabolism, and reactive oxygen metabolism, which are key in the occurrence and development of OA. There is evidence that drugs directly or indirectly targeting autophagy significantly hinder the progress of OA. In addition, the occurrence and development of autophagy in OA are regulated by many factors, including epigenetic modification, exosomes, crucial autophagy molecules, and signaling pathway regulation. Autophagy, as a new therapeutic target for OA, has widely influenced the pathological mechanism of OA. However, determining how autophagy affects OA pathology and its use in the treatment and diagnosis of targets still need further research.
Collapse
Affiliation(s)
- Chenglong Cheng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yuting Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Faxue Liao
- Department of Orthopaedics, The First Affiliated Hospital, Anhui Medical University, Hefei, China; Anhui Public Health Clinical Center, Hefei, China.
| | - Xiaomei Wang
- Department of Humanistic Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Institute of Rheumatism, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
44
|
Bordon G, Ramakrishna SN, Edalat SG, Eugster R, Arcifa A, Vermathen M, Aleandri S, Bertoncelj MF, Furrer J, Vermathen P, Isa L, Crockett R, Distler O, Luciani P. Liposomal aggregates sustain the release of rapamycin and protect cartilage from friction. J Colloid Interface Sci 2023; 650:1659-1670. [PMID: 37494862 DOI: 10.1016/j.jcis.2023.07.087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Liposomes show promise as biolubricants for damaged cartilage, but their small size results in low joint and cartilage retention. We developed a zinc ion-based liposomal drug delivery system for local osteoarthritis therapy, focusing on sustained release and tribological protection from phospholipid lubrication properties. Our strategy involved inducing aggregation of negatively charged liposomes with zinc ions to extend rapamycin (RAPA) release and improve cartilage lubrication. Liposomal aggregation occurred within 10 min and was irreversible, facilitating excess cation removal. The aggregates extended RAPA release beyond free liposomes and displayed irregular morphology influenced by RAPA. At nearly 100 µm, the aggregates were large enough to exceed the previously reported size threshold for increased joint retention. Tribological assessment on silicon surfaces and ex vivo porcine cartilage revealed the system's excellent protective ability against friction at both nano- and macro-scales. Moreover, RAPA was shown to attenuate the fibrotic response in human OA synovial fibroblasts. Our findings suggest the zinc ion-based liposomal drug delivery system has potential to enhance OA therapy through extended release and cartilage tribological protection, while also illustrating the impact of a hydrophobic drug like RAPA on liposome aggregation and morphology.
Collapse
Affiliation(s)
- Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Shivaprakash N Ramakrishna
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zurich, Vladimir- Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Sam G Edalat
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Remo Eugster
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Andrea Arcifa
- Laboratory for Surface Science and Coating Technologies, EMPA, Uberlandstrasse 129, 8600 Dubendorf, Switzerland
| | - Martina Vermathen
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | | | - Julien Furrer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Peter Vermathen
- Magnetic Resonance Methodology, Institute of Diagnostic and Interventional Neuroradiology, University & Inselspital Bern, sitem-insel AG, Freiburgstrasse 3, 3010 Bern, Switzerland
| | - Lucio Isa
- Laboratory for Soft Materials and Interfaces, Department of Materials, ETH Zurich, Vladimir- Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Rowena Crockett
- Laboratory for Surface Science and Coating Technologies, EMPA, Uberlandstrasse 129, 8600 Dubendorf, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| |
Collapse
|
45
|
Huang J, Chen Z, Wu Z, Xie X, Liu S, Kong W, Zhou J. Geniposide stimulates autophagy by activating the GLP-1R/AMPK/mTOR signaling in osteoarthritis chondrocytes. Biomed Pharmacother 2023; 167:115595. [PMID: 37769389 DOI: 10.1016/j.biopha.2023.115595] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 09/30/2023] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by cartilage degeneration. Autophagy is associated with chondrocyte homeostasis and exhibits a role in protecting against OA pathogenesis. Geniposide (GEN), an iridoid glycoside extracted from Eucommia ulmoides Oliv, acts as an activator of GLP-1R, which can stimulate autophagy. The AMPK/mTOR signaling pathway participates in the mediation of autophagy, and GLP-1R may act as an upstream factor of AMPK. However, whether GEN mediates the autophagic responses by activating the GLP-1R/AMPK/mTOR signaling pathway in OA chondrocytes is still unclear. In the current study, attenuated autophagy in MIA-induced rat OA models was observed, as shown by up-regulated expression of p62 and down-regulated expression of Beclin-1 and LC3-II/I. GEN stimulated autophagy and protected OA cartilage by up-regulating GLP-1R expression. In addition, GEN could enhance AMPK phosphorylation and down-regulate mTOR expression in IL-1β-treated C28/I2 cells. Inhibition of AMPK or activation of mTOR could reverse the stimulatory effects of GEN on autophagy. Furthermore, a GLP-1R inhibitor Exendin 9-39 could eliminate the chondroprotective effects of GEN by suppressing the AMPK/mTOR signaling pathway. Conclusively, Geniposide exhibits protective effects against osteoarthritis development by stimulating autophagy via activating the GLP-1R/AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jishang Huang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhenyu Wu
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Shiwei Liu
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
46
|
He X, Hu W, Zhang Y, Chen M, Ding Y, Yang H, He F, Gu Q, Shi Q. Cellular senescence in skeletal disease: mechanisms and treatment. Cell Mol Biol Lett 2023; 28:88. [PMID: 37891477 PMCID: PMC10612178 DOI: 10.1186/s11658-023-00501-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The musculoskeletal system supports the movement of the entire body and provides blood production while acting as an endocrine organ. With aging, the balance of bone homeostasis is disrupted, leading to bone loss and degenerative diseases, such as osteoporosis, osteoarthritis, and intervertebral disc degeneration. Skeletal diseases have a profound impact on the motor and cognitive abilities of the elderly, thus creating a major challenge for both global health and the economy. Cellular senescence is caused by various genotoxic stressors and results in permanent cell cycle arrest, which is considered to be the underlying mechanism of aging. During aging, senescent cells (SnCs) tend to aggregate in the bone and trigger chronic inflammation by releasing senescence-associated secretory phenotypic factors. Multiple signalling pathways are involved in regulating cellular senescence in bone and bone marrow microenvironments. Targeted SnCs alleviate age-related degenerative diseases. However, the association between senescence and age-related diseases remains unclear. This review summarises the fundamental role of senescence in age-related skeletal diseases, highlights the signalling pathways that mediate senescence, and discusses potential therapeutic strategies for targeting SnCs.
Collapse
Affiliation(s)
- Xu He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Wei Hu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Yuanshu Zhang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214026, People's Republic of China
| | - Mimi Chen
- Department of Orthopedics, Children Hospital of Soochow University, No. 92 Zhongnan Street, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Yicheng Ding
- Xuzhou Medical University, 209 Copper Mountain Road, Xuzhou, 221004, People's Republic of China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Fan He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
| | - Qiaoli Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214026, People's Republic of China.
| |
Collapse
|
47
|
Saengsiwaritt W, Ngamtipakon P, Udomsinprasert W. Vitamin D and autophagy in knee osteoarthritis: A review. Int Immunopharmacol 2023; 123:110712. [PMID: 37523972 DOI: 10.1016/j.intimp.2023.110712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Knee osteoarthritis (KOA), the highly prevalent degenerative disease affecting the joint, perpetually devastates the health of the elderly. Of various mechanisms known to participate in KOA etiology, apoptosis of chondrocytes is widely regarded as the primary cause of cartilage degradation. It has been suggested that the induction of autophagy in chondrocytes could potentially prolong the progression of KOA by modulating intracellular metabolic processes, which may be helpful for ameliorating chondrocyte apoptosis and eventual cartilage degeneration. Autophagy, a physiological process characterized by intracellular self-degradation, has been reportedly implicated in various pathologic conditions including KOA. Interestingly, vitamin D has been shown to regulate autophagy in human chondrocytes through multiple pathways, specifically AMPK/mTOR signaling pathway. This observation underscores the potential of vitamin D as a novel approach for restoring the functionality and survivability of chondrocytes in KOA. Supporting vitamin D's clinical significance, previous studies have demonstrated its substantial involvement in the symptoms and irregular joint morphology observed in KOA patients, strengthening potential therapeutic efficacy of vitamin D in treatment of KOA. Herein, the purpose of this review was to determine the mechanisms underlying the multi-processes of vitamin D implicated in autophagy in several cells including chondrocytes, which would bring unique insights into KOA pathogenesis.
Collapse
Affiliation(s)
| | - Phatchana Ngamtipakon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Wanvisa Udomsinprasert
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
48
|
Zhang Y, Liu C, Li Y, Xu H. Mechanism of the Mitogen-Activated Protein Kinases/Mammalian Target of Rapamycin Pathway in the Process of Cartilage Endplate Stem Cell Degeneration Induced by Tension Load. Global Spine J 2023; 13:2396-2408. [PMID: 35400210 PMCID: PMC10538332 DOI: 10.1177/21925682221085226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
STUDY DESIGN Basic Research. OBJECTIVE Intervertebral disc degeneration (IVDD) is caused by the cartilage endplate (CEP). Cartilage endplate stem cell (CESC) is involved in the recovery of CEP degeneration. Tension load (TL) contributes a lot to the initiation and progression of IVDD. This study aims to investigate the regulatory mechanism of the Mitogen-activated protein kinases/Mammalian target of rapamycin (MAPK/mTOR) pathway during TL-induced CESC degeneration. METHODS CESCs were isolated from New Zealand big-eared white female rabbits (6 months old). FX-4000T cell stress loading system was applied to establish a TL-induced degeneration model of CESCs. Western blotting was used to detect the level of mTOR pathway-related proteins and autophagy markers LC3-Ⅱ, Beclin-1, and p62 in degenerative CESCs. The expression of MAPK pathway-related proteins JNK and extracellular signal-regulated kinases (ERK) in degenerated CESCs was inhibited by cell transfection to explore whether JNK and ERK play a regulatory role in TL-induced autophagy in CESCs. RESULTS In the CESC degeneration model, the mTOR pathway was activated. After inhibition of mTOR, the autophagy level of CESCs was increased, and the degeneration of CESCs was alleviated. The MAPK pathway was also activated in the CESC degeneration model. Inhibition of JNK expression may alleviate TL-induced CEP degeneration by inhibiting Raptor phosphorylation and activating autophagy. Inhibition of ERK expression may alleviate TL-induced CEP degeneration by inhibiting mTOR phosphorylation and activating autophagy. CONCLUSION Inhibition of JNK and ERK in the MAPK signaling family alleviated TL-induced CESC degeneration by inhibiting the phosphorylation of Raptor and mTOR in the mTOR pathway.
Collapse
Affiliation(s)
- Yu Zhang
- Spine Research Center of Wannan Medical College, Department of Spine Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, P.R. China
| | - Chen Liu
- Spine Research Center of Wannan Medical College, Department of Spine Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, P.R. China
| | - Yu Li
- Spine Research Center of Wannan Medical College, Department of Spine Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, P.R. China
| | - Hongguang Xu
- Spine Research Center of Wannan Medical College, Department of Spine Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, P.R. China
| |
Collapse
|
49
|
Zhao S, Xiu G, Wang J, Wen Y, Lu J, Wu B, Wang G, Yang D, Ling B, Du D, Xu J. Engineering exosomes derived from subcutaneous fat MSCs specially promote cartilage repair as miR-199a-3p delivery vehicles in Osteoarthritis. J Nanobiotechnology 2023; 21:341. [PMID: 37736726 PMCID: PMC10515007 DOI: 10.1186/s12951-023-02086-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease involving cartilage. Exosomes derived from Mesenchymal stem cells (MSCs) therapy improves articular cartilage repair, but subcutaneous fat (SC) stromal cells derived exosomes (MSCsSC-Exos), especially engineering MSCsSC-Exos for drug delivery have been rarely reported in OA therapy. This objective of this study was to clarify the underlying mechanism of MSCsSC-Exos on cartilage repair and therapy of engineering MSCsSC-Exos for drug delivery in OA. MSCsSC-Exos could ameliorate the pathological severity degree of cartilage via miR-199a-3p, a novel molecular highly enriched in MSCsSC-Exos, which could mediate the mTOR-autophagy pathway in OA rat model. Intra-articular injection of antagomiR-199a-3p dramatically attenuated the protective effect of MSCsSC-Exos-mediated on articular cartilage in vivo. Furthermore, to achieve the superior therapeutic effects of MSCsSC-Exos on injured cartilage, engineering exosomes derived from MSCsSC as the chondrocyte-targeting miR-199a-3p delivery vehicles were investigated in vitro and in vivo. The chondrocyte-binding peptide (CAP) binding MSCsSC-Exos could particularly deliver miR-199a-3p into the chondrocytes in vitro and into deep articular tissues in vivo, then exert the excellent protective effect on injured cartilage in DMM-induced OA mice. As it is feasible to obtain human subcutaneous fat from healthy donors by liposuction operation in clinic, meanwhile engineering MSCsSC-Exos to realize targeted delivery of miR-199a-3p into chondrocytes exerted excellent therapeutic effects in OA animal model in vivo. Through combining MSCsSC-Exos therapy and miRNA therapy via an engineering approach, we develop an efficient MSCsSC-Exos-based strategy for OA therapy and promote the application of targeted-MSCsSC-Exos for drug delivery in the future.
Collapse
Affiliation(s)
- Shu Zhao
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120
- Department of Plastic Surgery, Shanghai Fourth People's Hospital, School of Medicine,Tongji University, Shanghai, 200434, People's Republic of China
| | - Guanghui Xiu
- Department of Intensive Care Unit, Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province), Yunnan University, Kunming, 650021, People's Republic of China
| | - Jian Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120
| | - Yi Wen
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120
| | - Jinyuan Lu
- Department of Hematology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Baitong Wu
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120
| | - Guangming Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120
| | - Danjing Yang
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120
| | - Bin Ling
- Department of Intensive Care Unit, Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province), Yunnan University, Kunming, 650021, People's Republic of China.
| | - Dajiang Du
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People's Republic of China.
| | - Jun Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China, 200120.
| |
Collapse
|
50
|
Yu Y, Lu S, Liu X, Li Y, Xu J. Identification and analysis of RNA-5-methylcytosine-related key genes in osteoarthritis. BMC Genomics 2023; 24:539. [PMID: 37700248 PMCID: PMC10496305 DOI: 10.1186/s12864-023-09651-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND 5-methylcytosine (m5C) modification is widely associated with many biological and pathological processes. However, knowledge of m5C modification in osteoarthritis (OA) remains lacking. Thus, our study aimed to identify common m5C features in OA. RESULTS In the present study, we identified 1395 differentially methylated genes (DMGs) and 1673 differentially expressed genes (DEGs) using methylated RNA immunoprecipitation next-generation sequencing (MeRIP-seq) and RNA-sequencing. A co-expression analysis of DMGs and DEGs showed that the expression of 133 genes was significantly affected by m5C methylation. A protein-protein interaction network of the 133 genes was constructed using the STRING database, and the cytoHubba plug-in of Cytoscape was used to hub genes were screen out 11 hub genes, including MMP14, VTN, COL15A1, COL6A2, SPARC, COL5A1, COL6A3, COL6A1, COL8A2, ADAMTS2 and COL7A1. The Pathway enrichment analysis by the ClueGO and CluePedia plugins in Cytoscape showed that the hub genes were significantly enriched in collagen degradation and extracellular matrix degradation. CONCLUSIONS Our study indicated that m5C modification might play an important role in OA pathogenesis, and the present study provides worthwhile insight into identifying m5C-related therapeutic targets in OA.
Collapse
Affiliation(s)
- Yang Yu
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shitao Lu
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoming Liu
- Department of Gastroenterology, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yu Li
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianzhong Xu
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|