1
|
Cai W, Wu A, Lin Z, Cao W, Pathak JL, Jaspers RT, Li R, Li X, Zheng K, Lin Y, Zhou N, Zhang X, Zhu Y, Zhang Q. S-propargyl-cysteine attenuates temporomandibular joint osteoarthritis by regulating macrophage polarization via Inhibition of JAK/STAT signaling. Mol Med 2025; 31:128. [PMID: 40197110 PMCID: PMC11974036 DOI: 10.1186/s10020-025-01186-6] [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: 12/18/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Temporomandibular joint osteoarthritis (TMJ-OA) is a disease characterized by cartilage degradation and synovial inflammation, with limited effective treatment currently. Synovial macrophage polarization is pivotal in TMJ-OA progression, making it a promising therapeutic aspect. This study investigated the effects of S-propargyl-cysteine (SPRC), an endogenous H2S donor, on macrophage polarization and its therapeutic potential in alleviating TMJ-OA. METHODS A MIA-induced TMJ-OA rat model and LPS-stimulated RAW264.7 macrophages were employed to evaluate the effects of SPRC in vivo and in vitro. TMJ bone and cartilage were analyzed via micro-CT and histological methods, while macrophage polarization markers expression were assessed via RT-qPCR, western blot, and immunofluorescence. RNA sequencing was performed on macrophages, and the JAK2/STAT3 signaling pathway was validated using the JAK2-specific inhibitor AG490. The direct effects of SPRC on rat primary condylar chondrocytes were examined by evaluating ECM synthesis and degradation. Co-culture experiments further assessed macrophage-chondrocyte interactions. RESULTS SPRC significantly alleviated cartilage and bone damage in the TMJ-OA rat model, as demonstrated by improved bone volume and cartilage structure. SPRC reduced pro-inflammatory M1 macrophage infiltration and enhanced anti-inflammatory M2 macrophage polarization. SPRC effectively inhibited the JAK2/STAT3, leading to reduction of inflammatory markers, including TNF-α, IL-6, and iNOS. Co-culture experiments revealed that SPRC-treated macrophage-conditioned medium improved chondrocyte metabolic activity and restored ECM integrity. CONCLUSIONS SPRC-modulated macrophage polarization alleviates TMJ-OA via JAK/STAT downregulation, thereby reducing synovial inflammation and cartilage degradation. These findings position SPRC as a promising therapeutic candidate for TMJ-OA and provide insights into novel strategies targeting macrophage polarization and synovium-cartilage crosstalk.
Collapse
Affiliation(s)
- Wenyi Cai
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Antong Wu
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Zhongxiao Lin
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wei Cao
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Janak L Pathak
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Richard T Jaspers
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Rui Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Xin Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Kaihan Zheng
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Yufu Lin
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Na Zhou
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
| | - Xin Zhang
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
| | - Yizhun Zhu
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China.
| | - Qingbin Zhang
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China.
| |
Collapse
|
2
|
Samad MA, Ahmad I, Hasan A, Alhashmi MH, Ayub A, Al‐Abbasi FA, Kumer A, Tabrez S. STAT3 Signaling Pathway in Health and Disease. MedComm (Beijing) 2025; 6:e70152. [PMID: 40166646 PMCID: PMC11955304 DOI: 10.1002/mco2.70152] [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: 09/30/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a critical transcription factor involved in multiple physiological and pathological processes. While STAT3 plays an essential role in homeostasis, its persistent activation has been implicated in the pathogenesis of various diseases, particularly cancer, bone-related diseases, autoimmune disorders, inflammatory diseases, cardiovascular diseases, and neurodegenerative conditions. The interleukin-6/Janus kinase (JAK)/STAT3 signaling axis is central to STAT3 activation, influencing tumor microenvironment remodeling, angiogenesis, immune evasion, and therapy resistance. Despite extensive research, the precise mechanisms underlying dysregulated STAT3 signaling in disease progression remain incompletely understood, and no United States Food and Drug Administration (USFDA)-approved direct STAT3 inhibitors currently exist. This review provides a comprehensive evaluation of STAT3's role in health and disease, emphasizing its involvement in cancer stem cell maintenance, metastasis, inflammation, and drug resistance. We systematically discuss therapeutic strategies, including JAK inhibitors (tofacitinib, ruxolitinib), Src Homology 2 domain inhibitors (S3I-201, STATTIC), antisense oligonucleotides (AZD9150), and nanomedicine-based drug delivery systems, which enhance specificity and bioavailability while reducing toxicity. By integrating molecular mechanisms, disease pathology, and emerging therapeutic interventions, this review fills a critical knowledge gap in STAT3-targeted therapy. Our insights into STAT3 signaling crosstalk, epigenetic regulation, and resistance mechanisms offer a foundation for developing next-generation STAT3 inhibitors with greater clinical efficacy and translational potential.
Collapse
Affiliation(s)
- Md Abdus Samad
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Iftikhar Ahmad
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Aakifah Hasan
- Department of BiochemistryFaculty of Life ScienceAligarh Muslim UniversityAligarhIndia
| | - Mohammad Hassan Alhashmi
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Medical Laboratory SciencesFaculty of Applied Medical SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Arusha Ayub
- Department of MedicineCollege of Health SciencesUniversity of GeorgiaGeorgiaUSA
| | - Fahad A. Al‐Abbasi
- Department of BiochemistryFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Ajoy Kumer
- Department of ChemistryCollege of Arts and SciencesInternational University of Business Agriculture & Technology (IUBAT)DhakaBangladesh
| | - Shams Tabrez
- King Fahd Medical Research CenterKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Medical Laboratory SciencesFaculty of Applied Medical SciencesKing Abdulaziz UniversityJeddahSaudi Arabia
| |
Collapse
|
3
|
Wang T, Zhao X, Li J, Yin C, Jiang B, Xie J, Wang B, Wang Y, Cao Z, Yao Q, Zheng S, Sui J, Zhu K. Oxytocin-loaded hydrogel promotes cartilage regeneration and regulates microenvironment. Biofabrication 2025; 17:025030. [PMID: 40096748 DOI: 10.1088/1758-5090/adc158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/17/2025] [Indexed: 03/19/2025]
Abstract
Osteoarthritis is a common orthopedic condition, and traditional treatment methods often fail to regenerate cartilage effectively. Oxytocin (OXT) is a neuropeptide that plays a crucial role in the skeletal system. Hyaluronic acid (HAMA) hydrogel has emerged as a key carrier for cartilage repair due to its excellent biocompatibility and biodegradability. Combining OXT with HAMA hydrogel and implanting it at the site of cartilage defects can effectively promote cartilage regeneration. Cartilage damage often results in an altered microenvironment, characterized by macrophage polarization and high levels of reactive oxygen species (ROS). Oxidative stress can stimulate macrophages to produce more pro-inflammatory factors. OXT can inhibit the secretion of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1βby interacting with the STAT3/NF-κB signaling pathway, as well as the PI3K/Akt and mitogen-activated protein kinase pathways, thereby inducing the polarization of macrophages from the M1 phenotype to the M2 phenotype and alleviating the inflammatory response. OXT can also enhance the expression of NRF and HO-1, which helps eliminate ROS and suppress the expression of pro-inflammatory factors. Regulating the microenvironment of cartilage damage is beneficial for cartilage protection and repair. OXT activates the CFOS/AP-1 and STAT1/JAK2 pathways, which together act on MMP2 and MMP9 to alleviate cartilage degeneration. The STAT1/JAK2 pathway can further increase the expression of Col2, thereby protecting chondrocytes. Additionally, OXT can directly boost the protein levels of SOX9 and COMP, promoting chondrocyte proliferation and cartilage protection, ultimately achieving the therapeutic goal for arthritis. This study explores the potential of HAMA hydrogel as a delivery system for OXT and analyzes their impact on cartilage regeneration and anti-inflammatory properties. This research provides a novel strategy for the treatment of cartilage injuries.
Collapse
Affiliation(s)
- Tianming Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Xiao Zhao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Jiayi Li
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Chongchong Yin
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Bo Jiang
- Children's Hospital of Nanjing Medical University, Nanjing 210000, People's Republic of China
| | - Jiaming Xie
- Children's Hospital of Nanjing Medical University, Nanjing 210000, People's Republic of China
| | - Binghao Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Yufeng Wang
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Zhicheng Cao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Qingqiang Yao
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Shengnai Zheng
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Jisheng Sui
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
| | - Kun Zhu
- Department of Orthopedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006 Nanjing, People's Republic of China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233000, People's Republic of China
| |
Collapse
|
4
|
Kaur B, Singh H, Choudhary G, Prakash A, Medhi B, Chatterjee D, Saini UC, Kaur J, Verma I, Sharma S. Natural angiotensin II type 1 receptor inhibitors: Virtual screening and in vitro evaluation of beta-1,2,3,4,6-penta-O-galloyl-d-glucopyranose, icarrin, and sesamin for osteoarthritis therapy. Int J Biol Macromol 2025; 309:142184. [PMID: 40147655 DOI: 10.1016/j.ijbiomac.2025.142184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025]
Affiliation(s)
- Bhavneet Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Harvinder Singh
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Gajendra Choudhary
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ajay Prakash
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Debajyoti Chatterjee
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Uttam Chand Saini
- Department of Orthopaedics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Indu Verma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Sadhna Sharma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| |
Collapse
|
5
|
Lu J, Shi Z, Geng L, Ren D, Hou H, Ren G, Yao S, Wang P. Transcriptional Analysis Reveals That the FHL1/JAK-STAT Pathway is Involved in Acute Cartilage Injury in Mice. Cartilage 2025:19476035251323601. [PMID: 40119525 PMCID: PMC11948231 DOI: 10.1177/19476035251323601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/24/2025] Open
Abstract
ObjectiveThis study aimed to identify genes and signaling pathways associated with acute cartilage injury using RNA sequencing (RNA-seq).MethodsKnee joint cartilage samples were collected from normal mice and 2 models of acute cartilage injury (non-invasive and groove models) within an 8-hour time limit. RNA-seq revealed differential gene expression between the injury models and controls, with subsequent validation using real-time quantitative polymerase chain reaction (RT-qPCR) for 9 representative genes.ResultsCompared to controls, the non-invasive model showed 36 differentially expressed genes (DEGs) (13 up-regulated, 23 down-regulated), with Gm14648 and Gm35438 showing the most significant upregulation and downregulation, respectively. The groove model exhibited 255 DEGs (13 up-regulated, 23 down-regulated), with Gm14648 and Gm35438 showing the (222 up-regulated, 33 down-regulated). Six overlapping genes were identified between the non-invasive and groove models, including up-regulated genes (Igfn1, Muc6, Hmox1) and down-regulated genes (Pthlh, Cyp1a1, Gm13490), validated by RT-qPCR. Gene ontology (GO) analysis highlighted involvement in environmental information processing and cartilage organ system function, while Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis implicated the JAK-STAT signaling pathway. RT-qPCR and immunohistochemistry confirmed downregulation of Fhl1 in the non-invasive model, supported by Western blotting of p-JAK2/t-JAK2 levels.ConclusionsThis study identifies DEGs (13 up-regulated, 23 down-regulated), with Gm14648 and Gm35438 showing the in acute cartilage injury, suggesting potential therapeutic targets. The role of Fhl1 in cartilage protection via the JAK-STAT pathway warrants further investigation in acute cartilage injury research.
Collapse
Affiliation(s)
- Jian Lu
- Department of Orthopedic Surgery, Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Zhenhua Shi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, People’s Republic of China
| | - Lindan Geng
- Department of Orthopedic Surgery, Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Dong Ren
- Department of Orthopedic Surgery, Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Haowei Hou
- Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Guowei Ren
- Department of Orthopedic Surgery, Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Shuangquan Yao
- Department of Orthopedic Surgery, Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Pengcheng Wang
- Department of Orthopedic Surgery, Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Orthopedic Research Institute of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| |
Collapse
|
6
|
庆 顺, 沈 智. [High expression of hexokinase 2 promotes proliferation, migration and invasion of colorectal cancer cells by activating the JAK/STAT pathway and regulating tumor immune microenvironment]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:542-553. [PMID: 40159969 PMCID: PMC11955892 DOI: 10.12122/j.issn.1673-4254.2025.03.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Indexed: 04/02/2025]
Abstract
OBJECTIVES To explore the expression of hexokinase 2 (HK2) in colorectal cancer (CRC) and its possible mechanisms for regulating tumor cell behaviors and tumor immune microenvironment. METHODS We analyzed HK2 expression in CRC and its impact on patient prognosis and tumor immune microenvironment using public databases. HK2 expression was also examined in 8 CRC and paired adjacent tissues using immunohistochemistry, Western blotting and RT-qPCR. In cultured CRC cell lines CT26 and HCT116 with low HK2 expression, the effects of lentivirus-mediated HK2 overexpression and JAK/STAT3 inhibitors on cell proliferation, migration, and invasion were assessed using CCK-8 assay, colony formation assay and Transwell assay and in a subcutaneous tumor-bearing mouse model; the changes were also observed in MC38 and CACO2 cells with high HK2 expressions following treatment with HK2 inhibitor 3-BP. Western blotting was performed to verify the relationship between HK2 and JAK/STAT signaling pathway protein expressions. RESULTS Informatics analyses suggested that HK2 expression was significantly higher in CRC tissues than in adjacent tissues (P<0.001), and patients with high HK2 expressions had worse prognosis (P=0.09). In the 8 clinical CRC tissues, HK2 expressions were significantly higher in the tumor tissues than in the adjacent tissues (P<0.01). In CT26 and HCT116 cells, HK2 overexpression significantly enhanced cell proliferation, migration and invasion, while in HK2-overexpressing MC38 and CACO2 cells, inhibiting HK2 with 3-BP strongly suppressed these changes. HK2 overexpression promoted STAT3 phosphorylation, and JAK/STAT3 inhibitors effectively suppressed tumor cell proliferation, migration and invasion. TIMER and MCPcounter analyses indicated correlations between HK2 and immune cells, and TCGA and GEO analyses suggested significant positive correlations between HK2 and the immune checkpoints including PDCD1. CONCLUSIONS HK2 is upregulated in CRC to promote tumor cell proliferation, migration and invasion possibly by activating the JAK-STAT signaling pathway and modulating tumor immune microenvironment.
Collapse
|
7
|
Alka, Mishra N, Singh P, Singh N, Rathore K, Verma V, Ratna S, Nisha R, Verma A, Saraf SA. Multifunctional polymeric nanofibrous scaffolds enriched with azilsartan medoxomil for enhanced wound healing. Drug Deliv Transl Res 2025; 15:846-873. [PMID: 38833068 DOI: 10.1007/s13346-024-01637-3] [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] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
A prolonged and compromised wound healing process poses a significant clinical challenge, necessitating innovative solutions. This research investigates the potential application of nanotechnology-based formulations, specifically nanofiber (NF) scaffolds, in addressing this issue. The study focuses on the development and characterization of multifunctional nanofibrous scaffolds (AZL-CS/PVA-NF) composed of azilsartan medoxomil (AZL) enriched chitosan/polyvinyl alcohol (CS/PVA) through electrospinning. The scaffolds underwent comprehensive characterization both in vitro and in vivo. The mean diameter and tensile strength of AZL-CS/PVA-NF were determined to be 240.42 ± 3.55 nm and 18.05 ± 1.18 MPa, respectively. A notable drug release rate of 93.86 ± 2.04%, was observed from AZL-CS/PVA-NF over 48 h at pH 7.4. Moreover, AZL-CS/PVA-NF exhibited potent antimicrobial efficacy for Staphylococcus aureus and Pseudomonas aeruginosa. The expression levels of Akt and CD31 were significantly elevated, while Stat3 showed a decrease, indicating a heightened tissue regeneration rate with AZL-CS/PVA-NF compared to other treatment groups. In vivo ELISA findings revealed reduced inflammatory markers (IL-6, IL-1β, TNF-α) within treated skin tissue, implying a beneficial effect on injury repair. The comprehensive findings of the present endeavour underscore the superior wound healing activity of the developed AZL-CS/PVA-NF scaffolds in a Wistar rat full-thickness excision wound model. This indicates their potential as novel carriers for drugs and dressings in the field of wound care.
Collapse
Affiliation(s)
- Alka
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
| | - Nidhi Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
| | - Priya Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
- School of Pharmacy, GITAM (Deemed-to-Be) University, Rudraram, Patancheru Mandal, Hyderabad, 502329, Telangana, India
| | - Neelu Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
| | - Kalpana Rathore
- Department of Materials Science and Engineering, Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India
| | - Vivek Verma
- Department of Materials Science and Engineering, Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India
- Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, 208016, Uttar Pradesh, India
- Samtel Centre for Display Technologies, Indian Institute of Technology Kanpur, 208016, Uttar Pradesh, India
- National Centre for Flexible Electronics, Indian Institute of Technology Kanpur, 208016, Uttar Pradesh, India
| | - Sheel Ratna
- Department of Environmental Microbiology, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
| | - Raquibun Nisha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
| | - Abhishek Verma
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India
| | - Shubhini A Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University Lucknow (A Central University), Uttar Pradesh, Vidya Vihar, Raebareli Road, Lucknow, 226025, Uttar Pradesh, India.
- National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, 226025, Uttar Pradesh, India.
| |
Collapse
|
8
|
Hosseini F, Ahmadi A, Sarvi ZN, Iranshahi M, Rassouli FB. 7-Geranyloxycoumarin modulated metastatic potential of osteosarcoma cells via interaction with MMPs and JAK1/2. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03847-z. [PMID: 39954065 DOI: 10.1007/s00210-025-03847-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/23/2025] [Indexed: 02/17/2025]
Abstract
Osteosarcoma (OS) is a highly aggressive bone cancer that primarily affects young adults. The tumor microenvironment and molecular mediators, including Janus kinases (JAKs) and matrix metalloproteinases (MMPs), significantly influence OS metastasis; activation of the JAK/STAT pathway enhances MMP expression and activity, promoting OS metastasis. 7-Geranyloxycoumarin, a natural agent found in various edible fruits and vegetables, possesses valuable pharmaceutical activities. This study aimed to investigate the effects of 7-geranyloxycoumarin on the metastasis of OS cells for the first time. To achieve this, a protein-protein interaction (PPI) network was constructed from the potential molecular and pathogenic targets associated with 7-geranyloxycoumarin and OS to identify overlapping targets. Subsequently, GO and KEGG pathway enrichment analyses were conducted. Molecular docking and dynamic simulations were also performed to elucidate the binding affinity of 7-geranyloxycoumarin with JAK1 and JAK2. For in vitro studies, 7-geranyloxycoumarin was first extracted from Ferula szowitsiana using thin-layer chromatography. The cells were then treated and evaluated for viability, apoptosis, migration, invasion, adhesion, and MMPs activity. The study identified 50 shared targets and revealed MMP-2, MMP-9, JAK1, and JAK2 as hub genes, confirmed through enrichment analyses. Molecular docking revealed strong interactions between 7-geranyloxycoumarin and JAK1 and JAK2 proteins, and molecular dynamics simulations indicated both conformational flexibility and binding stability of the ligand-protein complex. Moreover, experimental studies demonstrated that 7-geranyloxycoumarin did not induce apoptosis but significantly altered the migration, invasion, and adhesion of OS cells by inhibiting the activity of MMPs. In conclusion, 7-geranyloxycoumarin is proposed as a promising therapeutic agent for targeting metastasis in OS cells.
Collapse
Affiliation(s)
- Fatemehsadat Hosseini
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abdolreza Ahmadi
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zahra Nasiri Sarvi
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
9
|
Chun JM, Park JH, Moon BC, Baek SJ. Transcriptomic insights into the anti-inflammatory mechanisms of Protaetia brevitarsis seulensis larvae in IL-1β-driven chondrosarcoma cells. Biomed Pharmacother 2025; 183:117866. [PMID: 39862704 DOI: 10.1016/j.biopha.2025.117866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Osteoarthritis (OA) is a complex, degenerative, multi-factorial joint disease. Because of the difficulty in treating OA, developing new targeting strategies that can be used to understand its molecular mechanisms is critical. Protaetia brevitarsis seulensis larvae offer much therapeutic value; however, the presence of various active compounds and the multi-factorial risk factors for OA render the precise mechanisms of action unclear. A systematic transcriptome analysis was used to investigate the key mechanisms of action of P. brevitarsis seulensis larvae aqueous extract (PBSL) and its compounds on OA. Major mechanisms and transcription factors of PBSL were analyzed by profiling gene expression changes in interleukin (IL)-1β-induced human chondrosarcoma cell (SW1353) treated with PBSL. An in vitro assay was performed to validate the efficacy of the novel mechanism and targets of PBSL. PBSL exerted anti-inflammatory effects on SW1353 cells by regulating many molecular pathways. The IL-6/JAK/STAT3 pathway was significantly downregulated by PBSL, and STAT3 was identified as a major transcription factor regulating PBSL-induced target gene expression. Of the six PBSL compounds, the major compound was regulated by the IL-6/JAK/STAT3 pathway. This study provided potential novel mechanisms and transcription factors for PBSL and its active compounds against OA and indicated that inhibiting the IL-6/JAK/STAT3 pathway is a therapeutic target for treating OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Digital Health Research Division, Korean Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korean Institute of Oriental Medicine, Naju 58245, Republic of Korea
| | - Byeong Cheol Moon
- Herbal Medicine Resources Research Center, Korean Institute of Oriental Medicine, Naju 58245, Republic of Korea
| | - Su-Jin Baek
- Korean Medicine Data Division, Korean Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
10
|
Ning P, Lin S, Shi Y, Liu T. Potential role of gut-related factors in the pathology of cartilage in osteoarthritis. Front Nutr 2025; 11:1515806. [PMID: 39845920 PMCID: PMC11753001 DOI: 10.3389/fnut.2024.1515806] [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: 10/23/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Osteoarthritis (OA) is a common progressive degenerative disease. Gut microbiota (GM) and their metabolites have been closely associated with the onset, progression, and pathology of OA. GM and their metabolites may influence the cartilage directly, or indirectly by affecting the gut, the immune system, and the endocrine system. They function through classical pathways in cartilage metabolism and novel pathways that have recently been discovered. Some of them have been used as targets for the prevention and treatment of OA. The current study sought to describe the major pathological signaling pathways in OA chondrocytes and the potential role of gut-related factors in these pathways.
Collapse
Affiliation(s)
- Peng Ning
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuting Lin
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongyan Shi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianjing Liu
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Jia J, Zhou X, Chu Q. Mechanisms and therapeutic prospect of the JAK-STAT signaling pathway in liver cancer. Mol Cell Biochem 2025; 480:1-17. [PMID: 38519710 DOI: 10.1007/s11010-024-04983-5] [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: 12/12/2023] [Accepted: 02/29/2024] [Indexed: 03/25/2024]
Abstract
Liver cancer (LC) poses a significant global health challenge due to its high incidence and poor prognosis. Current systemic treatment options, such as surgery, chemotherapy, radiofrequency ablation, and immunotherapy, have shown limited effectiveness for advanced LC patients. Moreover, owing to the heterogeneous nature of LC, it is crucial to uncover more in-depth pathogenic mechanisms and develop effective treatments to address the limitations of the existing therapeutic modalities. Increasing evidence has revealed the crucial role of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in the pathogenesis of LC. The specific mechanisms driving the JAK-STAT pathway activation in LC, participate in a variety of malignant biological processes, including cell differentiation, evasion, anti-apoptosis, immune escape, and treatment resistance. Both preclinical and clinical investigations on the JAK-STAT pathway inhibitors have exhibited potential in LC treatment, thereby opening up avenues for the development of more targeted therapeutic strategies for LC. In this study, we provide an overview of the JAK-STAT pathway, delving into the composition, activation, and dynamic interplay within the pathway. Additionally, we focus on the molecular mechanisms driving the aberrant activation of the JAK-STAT pathway in LC. Furthermore, we summarize the latest advancements in targeting the JAK-STAT pathway for LC treatment. The insights presented in this review aim to underscore the necessity of research into the JAK-STAT signaling pathway as a promising avenue for LC therapy.
Collapse
Affiliation(s)
- JunJun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China.
| | - Xuelian Zhou
- Division of Endocrinology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
12
|
Yang X, Li J, Xu C, Zhang G, Che X, Yang J. Potential mechanisms of rheumatoid arthritis therapy: Focus on macrophage polarization. Int Immunopharmacol 2024; 142:113058. [PMID: 39236455 DOI: 10.1016/j.intimp.2024.113058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that affects multiple organs and systems in the human body, often leading to disability. Its pathogenesis is complex, and the long-term use of traditional anti-rheumatic drugs frequently results in severe toxic side effects. Therefore, the search for a safer and more effective antirheumatic drug is extremely important for the treatment of RA. As important immune cells in the body, macrophages are polarized. Under pathological conditions, macrophages undergo proliferation and are recruited to diseased tissues upon stimulation. In the local microenvironment, they polarize into different types of macrophages in response to specific factors and perform unique functions and roles. Previous studies have shown that there is a link between macrophage polarization and RA, indicating that certain active ingredients can ameliorate RA symptoms through macrophage polarization. Notably, Traditional Chinese medicine (TCM) monomer component and compounds demonstrate a particular advantage in this process. Building upon this insight, we reviewed and analyzed recent studies to offer valuable and meaningful insights and directions for the development and application of anti-rheumatic drugs.
Collapse
Affiliation(s)
- Xinyu Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinling Li
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chengchao Xu
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinzhen Che
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
13
|
Altahla R, Alshorman J, Tao X. Ferroptosis plays a role in osteoarthritis. ALL LIFE 2024; 17. [DOI: 10.1080/26895293.2024.2391292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/28/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Ruba Altahla
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jamal Alshorman
- Department of Orthopedics, The Second Affiliated Hospital, Hubei University of Science and Technology, Xiang Ning, People’s Republic of China
| | - Xu Tao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
14
|
Shang Y, Zhao K, Xue W, An J, Zhong Y, Chen Y, Zeng Q, Tang Q, Qiu X. Comparative assessment of acute neurotoxicity of real-world ultra-fine black carbon emitted from residential solid fuel combustion. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176597. [PMID: 39349200 DOI: 10.1016/j.scitotenv.2024.176597] [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: 07/15/2024] [Revised: 09/16/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
Incomplete combustion of residential solid fuel is one of the main anthropogenic sources for black carbon (BC). Fresh BC, mainly enriched in ultra-fine fraction of particles, can directly cross blood-brain barrier and are reported to be associated with neurodegenerative diseases. Because of the difficulties in collection and purification of BC from ambient particles, there are still significant knowledge gaps in understanding neurotoxicity caused by real-world BC. The purpose of this study is to compare the neurotoxic effects caused by BCs emitted from combustion of six residential solid fuels, and try to reveal associated biological mechanisms in SH-SY5Y cells. Two straw BC (Wheat-BC and Corn-BC) showed highest neurotoxic effects followed by wood BC (Pine-BC and Aspen-BC) and coal BC (Xvzhou and Longkou Coal), as indicated by viability, lactic dehydrogenase, malondialdehyde, adenosine triphosphate and acetylcholine levels. Coal BC caused nearly no toxicity in human neuroblastoma (SH-SY5Y) cells within highest dose of 200 μg/mL. RNA sequence and bioinformatics analysis were applied to effectively identify differential genes and signaling pathways. Based on Gene Ontology (GO) annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, Protein Protein Interaction network (PPI network) construction, we found biomass BC affected mitochondrial function, interfered with cellular metabolic processes, disturbed redox homeostasis, and finally resulted in cellular damages. Coal-BC mainly caused cytokine/chemokine related inflammatory responses. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting methods were further applied to find out related signaling pathways. Biomass BC activated IL6R/JAK3/STAT3 and JAK3/STAT6 pathways leading to oxidative stress and inflammatory responses. Coal BC activated JAK3/STAT3 pathway leading to chemokine related responses. This study revealed the heterogeneity in neurotoxicity of BCs from different combustion sources and provided important data for health risk assessment. BC-related neurotoxicity should be considered when making air pollution emission control strategies, with residential biomass receiving more policy attention.
Collapse
Affiliation(s)
- Yu Shang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Kunming Zhao
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Wanlei Xue
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Jing An
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yufang Zhong
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yingjun Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Qingming Zeng
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Qisheng Tang
- Fudan University Huashan Hospital, Department of Neurosurgery, National Center for Neurological Disorders, National Key Laboratory for Medical Neurobiology, Shanghai Key Laboratory of Brain Function and Regeneration, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Shanghai Medical College-Fudan University, Shanghai 200040, China.
| | - Xinghua Qiu
- State Key Joint Laboratory for Environmental Simulation and Pollution Control, State Environmental Protection Key Laboratory of Atmospheric Exposure and Health Risk Management, College of Environmental Sciences and Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
15
|
Ruan S, Tang D, Luo Y, Song H. Identification of mitophagy-related biomarkers in osteoarthritis. Animal Model Exp Med 2024; 7:781-792. [PMID: 38720455 PMCID: PMC11680475 DOI: 10.1002/ame2.12416] [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: 01/16/2024] [Accepted: 03/27/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common joint disease, and existing drugs cannot cure OA, so there is an urgent need to identify new targets. Mitophagy plays an important role in OA; however, the role of mitophagy in the OA immune system is not yet clear. METHODS In this study, differential analysis and enrichment analysis were used to identify mitophagy-related genes (MRGs) with differential expression in OA and the functional pathways involved in OA. Subsequently, two machine learning methods, RF and LASSO, were used to screen MRGs with diagnostic value and construct nomograms. At the same time, the relationship between mitophagy and OA immune response was explored by immunoinfiltration analysis. RESULTS Forty-three differentially MRGs were identified in OA, of which six MRGs (GABARAPL2, PARL, GABARAPL1, JUN, RRAS, and SNX7) were associated with the diagnosis of OA. The ROC analysis results show that these 6 MRGs have high predictive accuracy in the diagnosis of OA. In immune infiltration analysis, we found that the abundance of significantly different immune cells in OA was mostly upregulated. In addition, the expression of diagnostic-related MRGs is correlated with changes in the abundance of immune cells in OA. CONCLUSION This study demonstrates that six MRGs can be used as diagnostic biomarkers. The expression of diagnostic-related MRGs is correlated with changes in the abundance of immune cells in OA. At the same time, mitophagy may affect the immune microenvironment of OA by regulating immune cells, ultimately leading to the progression of OA.
Collapse
Affiliation(s)
- Shiqiang Ruan
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| | - Dongxu Tang
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| | - Yanfei Luo
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| | - Hao Song
- Department of Orthopaedics Surgerythe First People's Hospital of Zunyi City (The Third Affiliated Hospital of Zunyi Medical University)ZunyiChina
| |
Collapse
|
16
|
Zeng L, Wang Y, Shen J, Wei X, Wu Y, Chi X, Zheng X, Yu X, Shi Y, Liu W. TIPE2 aggravates experimental colitis and disrupts intestinal epithelial barrier integrity by activating JAK2/STAT3/SOCS3 signal pathway. Exp Cell Res 2024; 443:114287. [PMID: 39426612 DOI: 10.1016/j.yexcr.2024.114287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Ulcerative colitis (UC) is a chronic relapsing and progressive inflammatory disease of the colon. TIPE2 is a negative regulator of innate and adaptive immunity that maintains immune homeostasis. We found that TIPE2 was highly expressed in mucosa of mice with colitis. However, the role of TIPE2 in colitis remains unclear. We induced colitis in mice with dextran sulfate sodium (DSS) and treated them with TIPE2, and investigated the inflammatory activity of the colon in vivo by cytokines detection and histopathological analyses. We also measured inflammatory alteration and tight junctions induced by DSS in vitro. The results demonstrated that administration of TIPE2 promoted the severity of colitis in mice and human colon epithelial cells. Furthermore, TIPE2 aggravated intestinal epithelial barrier dysfunction by decreasing the expression of the tight junction proteins Occludin, Claudin-1 and ZO-1. In addition, TIPE2 exacerbated intestinal inflammatory response by inhibiting the expression of SOCS3, remarkably activating JAK2/STAT3 signaling pathway, and increasing the translocation of phosphorylated STAT3 into the nucleus. Silencing of TIPE2 attenuated the DSS-induced activation of JAK2/STAT3, thereby rescuing epithelial inflammatory injury and restoring barrier dysfunction. These results indicate that TIPE2 augments experimental colitis and disrupted the integrity of the intestinal epithelial barrier by activating the JAK2/STAT3/SOCS3 signaling pathway.
Collapse
Affiliation(s)
- Lingli Zeng
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuping Wang
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiaxin Shen
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xujin Wei
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yilong Wu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xintong Chi
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xueyan Zheng
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xing Yu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Shi
- Department of Gastroenterology, The First Afiiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Wenming Liu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
17
|
Li Y, Wang GQ, Li YB. Therapeutic potential of natural coumarins in autoimmune diseases with underlying mechanisms. Front Immunol 2024; 15:1432846. [PMID: 39544933 PMCID: PMC11560467 DOI: 10.3389/fimmu.2024.1432846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Autoimmune diseases encompass a wide range of disorders characterized by disturbed immunoregulation leading to the development of specific autoantibodies, which cause inflammation and multiple organ involvement. However, its pathogenesis remains unelucidated. Furthermore, the cumulative medical and economic burden of autoimmune diseases is on the rise, making these diseases a ubiquitous global phenomenon that is predicted to further increase in the coming decades. Coumarins, a class of aromatic natural products with benzene and alpha-pyrone as their basic structures, has good therapeutic effects on autoimmune diseases. In this review, we systematically highlighted the latest evidence on coumarins and autoimmune diseases data from clinical and animal studies. Coumarin acts on immune cells and cytokines and plays a role in the treatment of autoimmune diseases by regulating NF-κB, Keap1/Nrf2, MAPKs, JAK/STAT, Wnt/β-catenin, PI3K/AKT, Notch and TGF-β/Smad signaling pathways. This systematic review will provide insight into the interaction of coumarin and autoimmune diseases, and will lay a groundwork for the development of new drugs for autoimmune diseases.
Collapse
Affiliation(s)
- Yan Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Shandong Provincial Key Medical and Health Laboratory of Neuroimmunology, Jinan, China
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guan-qing Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Shandong Provincial Key Medical and Health Laboratory of Neuroimmunology, Jinan, China
| | - Yan-bin Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Shandong Provincial Key Medical and Health Laboratory of Neuroimmunology, Jinan, China
| |
Collapse
|
18
|
Zhang Y, Tao H, Zhang L, Li X, Shi Y, Sun W, Chen W, Zhao Y, Wang L, Yang X, Gu C. Battling pain from osteoarthritis: causing novel cell death. Acta Biochim Biophys Sin (Shanghai) 2024; 57:169-181. [PMID: 39463202 PMCID: PMC11877141 DOI: 10.3724/abbs.2024189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Osteoarthritis (OA) is a significant contributor to pain and disability worldwide. Pain is the main complaint of OA patients attending the clinic and has a large impact on their quality of life and economic standards. However, existing treatments for OA-related pain have not been shown to achieve good relief. The main focus is on preventing and slowing the progression of OA so that the problem of OA pain can be resolved. Pain caused by OA is complex, with the nature, location, duration, and intensity of pain changing as the disease progresses. Previous research has highlighted the role of various forms of cell death, such as apoptosis and necrosis, in the progression of pain in OA. Emerging studies have identified additional forms of novel cell death, such as pyroptosis, ferroptosis, and necroptosis that are linked to pain in OA. Different types of cell death contribute to tissue damage in OA by impacting inflammatory responses, reactive oxygen species (ROS) production, and calcium ion levels, ultimately leading to the development of pain. Evidence suggests that targeting novel types of cell death could help alleviate pain in OA patients. This review delves into the complex mechanisms of OA pain, explores the relationship between different modes of novel cell death and pain, and proposes novel cell death as a viable strategy for the treatment of these conditions, with the goal of providing scientific references for the development of future OA pain treatments and drugs.
Collapse
Affiliation(s)
- Yuheng Zhang
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Huaqiang Tao
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversitySuzhou226000China
| | - Liyuan Zhang
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Xueyan Li
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Yi Shi
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Wen Sun
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Wenlong Chen
- Orthopedics and Sports Medicine CenterSuzhou Municipal HospitalNanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Yuhu Zhao
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversitySuzhou226000China
| | - Liangliang Wang
- Department of Orthopedicsthe Affiliated Changzhou Second People’s Hospital of
Nanjing Medical UniversityChangzhou213003China
| | - Xing Yang
- Orthopedics and Sports Medicine CenterSuzhou Municipal HospitalNanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| | - Chengyong Gu
- Anesthesiology DepartmentSuzhou Municipal Hospital (North District)Nanjing Medical University Affiliated Suzhou HospitalSuzhou226000China
| |
Collapse
|
19
|
Cho SW, Sontam T, Batta S, Parekh PK. Alopecia areata is associated with an increased risk of osteoarthritis of the knee and hip: a retrospective cohort analysis. Arch Dermatol Res 2024; 316:672. [PMID: 39387916 DOI: 10.1007/s00403-024-03386-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 08/27/2024] [Accepted: 09/14/2024] [Indexed: 10/12/2024]
Affiliation(s)
- Seo Won Cho
- Texas A&M College of Medicine, Dallas, Texas, USA
| | - Tarun Sontam
- Texas A&M College of Medicine, Dallas, Texas, USA
| | - Sonali Batta
- Department of Dermatology, Baylor Scott & White Health, Temple, Texas, USA
| | - Palak K Parekh
- Department of Dermatology, Baylor Scott & White Health, Temple, Texas, USA.
| |
Collapse
|
20
|
Wang S, Kurth S, Burger C, Wirtz DC, Schildberg FA, Ossendorff R. TNFα-Related Chondrocyte Inflammation Models: A Systematic Review. Int J Mol Sci 2024; 25:10805. [PMID: 39409134 PMCID: PMC11476358 DOI: 10.3390/ijms251910805] [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: 08/05/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
Tumor necrosis factor alpha (TNFα), as a key pro-inflammatory cytokine, plays a central role in joint diseases. In recent years, numerous models of TNFα-induced cartilage inflammation have been developed. However, due to the significant differences between these models and the lack of consensus in their construction, it becomes difficult to compare the results of different studies. Therefore, we summarized and compared these models based on important parameters for model construction, such as cell source, cytokine concentration, stimulation time, mechanical stimulation, and more. We attempted to analyze the advantages and disadvantages of each model and provide a compilation of the analytical methods used in previous studies. Currently, TNFα chondrocyte inflammation models can be categorized into four main types: monolayer-based, construct-based, explant-based TNFα chondrocyte inflammation models, and miscellaneous TNFα chondrocyte inflammation models. The most commonly used models were the monolayer-based TNFα chondrocyte inflammation models (42.86% of cases), with 10 ng/mL TNFα being the most frequently used concentration. The most frequently used chondrocyte cell passage is passage 1 (50%). Human tissues were most frequently used in experiments (51.43%). Only five articles included models with mechanical stimulations. We observed variations in design conditions between different models. This systematic review provides the essential experimental characteristics of the available chondrocyte inflammation models with TNFα, and it provides a platform for better comparison between existing and new studies in this field. It is essential to perform further experiments to standardize each model and to find the most appropriate experimental parameters.
Collapse
|
21
|
Chen W, Xiao J, Zhou Y, Liu W, Jian J, Yang J, Chen B, Ye Z, Liu J, Xu X, Jiang T, Wang H, Liu W. Curcumenol regulates Histone H3K27me3 demethylases KDM6B affecting Succinic acid metabolism to alleviate cartilage degeneration in knee osteoarthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155922. [PMID: 39126921 DOI: 10.1016/j.phymed.2024.155922] [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: 04/30/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Cartilage metabolism dysregulation is a crucial driver in knee osteoarthritis (KOA). Modulating the homeostasis can mitigate the cartilage degeneration in KOA. Curcumenol, derived from traditional Chinese medicine Curcuma Longa L., has demonstrated potential in enhancing chondrocyte proliferation and reducing apoptosis. However, the specific mechanism of Curcumenol in treating KOA remains unclear. This study aimed to demonstrate the molecular mechanism of Curcumenol in treating KOA based on the transcriptomics and metabolomics, and both in vivo and in vitro experimental validations. MATERIALS AND METHODS In this study, a destabilization medial meniscus (DMM)-induced KOA mouse model was established. And the mice were intraperitoneally injected with Curcumenol at 4 and 8 mg/kg concentrations. The effects of Curcumenol on KOA cartilage and subchondral was evaluated using micro-CT, histopathology, and immunohistochemistry (IHC). In vitro, OA chondrocytes were induced with 10 μg/mL lipopolysaccharide (LPS) and treated with Curcumenol to evaluate the proliferation, apoptosis, and extracellular matrix (ECM) metabolism through CCK8 assay, flow cytometry, and chondrocyte staining. Furthermore, transcriptomics and metabolomics were utilized to identify differentially expressed genes (DEGs) and metabolites. Finally, integrating multi-omics analysis, virtual molecular docking (VMD), and molecular dynamics simulation (MDS), IHC, immunofluorescence (IF), PCR, and Western blot (WB) validation were conducted to elucidate the mechanism by which Curcumenol ameliorates KOA cartilage degeneration. RESULTS Curcumenol ameliorated cartilage destruction and subchondral bone loss in KOA mice, promoted cartilage repair, upregulated the expression of COL2 while downregulated MMP3, and improved ECM synthesis metabolism. Additionally, Curcumenol also alleviated the damage of LPS on the proliferation activity and suppressed apoptosis, promoted ECM synthesis. Transcriptomic analysis combined with weighted gene co-expression network analysis (WGCNA) identified a significant downregulation of 19 key genes in KOA. Metabolomic profiling showed that Curcumenol downregulates the expression of d-Alanyl-d-alanine, 17a-Estradiol, Glutathione, and Succinic acid, while upregulating Sterculic acid and Azelaic acid. The integrated multi-omics analysis suggested that Curcumenol targeted KDM6B to regulate downstream protein H3K27me3 expression, which inhibited methylation at the histone H3K27, consequently reducing Succinic acid levels and improving KOA cartilage metabolism homeostasis. Finally, both in vivo and in vitro findings indicated that Curcumenol upregulated KDM6B, suppressed H3K27me3 expression, and stimulated collagen II expression and ECM synthesis, thus maintaining cartilage metabolism homeostasis and alleviating KOA cartilage degeneration. CONCLUSION Curcumenol promotes cartilage repair and ameliorates cartilage degeneration in KOA by upregulating KDM6B expression, thereby reducing H3K27 methylation and downregulating Succinic Acid, restoring metabolic stability and ECM synthesis.
Collapse
Affiliation(s)
- Weijian Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Jiacong Xiao
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Yi Zhou
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Weinian Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Junde Jian
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510045, Guangdong, China
| | - Jiyong Yang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Bohao Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Zhilong Ye
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Jun Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Xuemeng Xu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China
| | - Tao Jiang
- Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China.
| | - Haibin Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China; Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China.
| | - Wengang Liu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510095, Guangdong, China; Guangdong Provincial Second Hospital of Traditional Chinese Medicine (Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, Guangdong, China; Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou 510095, Guangdong, China.
| |
Collapse
|
22
|
Zhang Z, Mao H, Li F, Wang D, Liu Y. METTL14-mediated lncRNA-FAS-AS1 promotes osteoarthritis progression by up-regulating ADAM8. Int J Rheum Dis 2024; 27:e15323. [PMID: 39221886 DOI: 10.1111/1756-185x.15323] [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: 04/26/2024] [Revised: 08/02/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent degenerative disease. We explored the role and regulatory mechanisms of lncRNA-FAS-AS1 in OA progression. METHODS We exposed human immortalized chondrocytes to IL-1β for 24 h to induce an OA cell model. The target molecule levels were assessed using western blot and quantitative real-time PCR (RT-qPCR). Cell viability and apoptosis were measured using CCK-8 and flow cytometry. The m6A modification of FAS-AS1 was determined using MeRIP. We examined the binding relationships between FAS-AS1, Fragile X mental retardation 1 (FMR1), and A disintegrin and metalloproteinase 8 (ADAM8) using RIP and RNA pull-down. The OA animal model was established by separating the medial collateral ligament and medial meniscus. Safranin-O staining and Mankin's scale were employed to evaluate pathological changes within the cartilage. RESULTS FAS-AS1, METTL14, and ADAM8 were upregulated, and the JAK/STAT3 signaling pathway was activated in OA mice and IL-1β-induced chondrocytes. FAS-AS1 knockdown inhibited extracellular matrix degradation in IL-1β-induced chondrocytes; however, ADAM8 overexpression reversed this effect. FAS-AS1 maintained the stability of ADAM8 mRNA by recruiting FMR1. METTL14 knockdown repressed FAS-AS1 expression in an m6A-dependent manner. FAS-AS1 overexpression reversed the inhibitory effects of METTL14 knockdown on JAK/STAT3 signaling and cartilage damage in the OA model both in vitro and in vivo. CONCLUSION METTL14-mediated FAS-AS1 promotes OA progression through the FMR1/ADAM8/JAK/STAT3 axis.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- ADAM Proteins/metabolism
- ADAM Proteins/genetics
- Adenosine/analogs & derivatives
- Apoptosis
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cell Line
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Disease Models, Animal
- Disease Progression
- Interleukin-1beta/metabolism
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Mice, Inbred C57BL
- Osteoarthritis/metabolism
- Osteoarthritis/genetics
- Osteoarthritis/pathology
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Signal Transduction
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Up-Regulation
Collapse
Affiliation(s)
- Zhehua Zhang
- Department of Orthopedics, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia BaoGang Hospital, Baotou, Inner Mongolia, China
| | - Honggang Mao
- Department of Orthopedics, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia BaoGang Hospital, Baotou, Inner Mongolia, China
| | - Fang Li
- Department of Experimental Center, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia BaoGang Hospital, Baotou, Inner Mongolia, China
| | - Dahai Wang
- Department of Orthopedics, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia BaoGang Hospital, Baotou, Inner Mongolia, China
| | - Yan Liu
- Department of Orthopedics, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia BaoGang Hospital, Baotou, Inner Mongolia, China
| |
Collapse
|
23
|
Zhao W, Zhu Y, Wong SK, Muhammad N, Pang KL, Chin KY. Effects of resveratrol on biochemical and structural outcomes in osteoarthritis: A systematic review and meta-analysis of preclinical studies. Heliyon 2024; 10:e34064. [PMID: 39055794 PMCID: PMC11269911 DOI: 10.1016/j.heliyon.2024.e34064] [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: 11/04/2023] [Revised: 05/22/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
Background and objective Osteoarthritis (OA) is the most common age-related disease of joints with increasing global prevalence. Persistent inflammation within the joint space is speculated to be the cause of OA. Resveratrol is an anti-inflammatory and antioxidant compound which can influence cartilage metabolism through multiple signalling pathways. This systematic review and meta-analysis aimed to summarize the therapeutic effects of resveratrol in animal models of OA. Methods A comprehensive literature search was performed using PubMed, Embase, Web of Science, Cochrane Library, China National Knowledge Infrastructure, China Wanfang and VIP databases in May 2023. Studies on the effects of resveratrol in animal models of OA written in English or Mandarin, published from the inception of databases until the date of the search were considered. Results Fifteen eligibility studies were included and analysed. Resveratrol was shown to inhibit the secretion of interleukin-1β, tumour necrosis factor-α, interleukin-6, nitric oxide, and apoptosis of articular chondrocytes. Joint structure as indicated by Mankin scores was restored with resveratrol in animal OA models. Conclusion Resveratrol is a potential therapeutic agent for OA based on animal studies. Further evidence from well-planned human studies would be required to validate its clinical efficacies.
Collapse
Affiliation(s)
- Wenjian Zhao
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Malaysia
- Department of Pathology, College of Basic Medicine, Xiangnan University, 423000, Chenzhou City, China
| | - Yuezhi Zhu
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Malaysia
| | - Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Malaysia
| | - Norliza Muhammad
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Malaysia
| | - Kok-Lun Pang
- Newcastle University Medicine Malaysia, 79200, Iskandar Puteri, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Cheras, Malaysia
| |
Collapse
|
24
|
Fazio A, Di Martino A, Brunello M, Traina F, Marvi MV, Mazzotti A, Faldini C, Manzoli L, Evangelisti C, Ratti S. The involvement of signaling pathways in the pathogenesis of osteoarthritis: An update. J Orthop Translat 2024; 47:116-124. [PMID: 39021400 PMCID: PMC11254498 DOI: 10.1016/j.jot.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 04/09/2024] [Accepted: 06/02/2024] [Indexed: 07/20/2024] Open
Abstract
Osteoarthritis (OA) is one of the most common disabling pathologies, characterized by joint pain and reduced function, significantly worsening the quality of life. Even if important progresses have been made in OA research, little is yet known about the precise cellular and molecular mechanisms underlying OA. Understanding dysregulated signaling networks and their crosstalk in OA may offer a strong opportunity for the development of combined targeted therapies. Hence, this review highlights the recent findings on the main pathways involved in OA development, including Wnt, Notch, Hedgehog, MAPK, AMPK, and JAK/STAT, providing insights on current targeted therapies in OA patients' management. The translational potential of this article The identification of key signaling pathways involved in OA development and the investigation of their signaling crosstalk could pave the way for more effective treatments and improved management of OA patients in the future.
Collapse
Affiliation(s)
- Antonietta Fazio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Alberto Di Martino
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Matteo Brunello
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Francesco Traina
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ortopedia-Traumatologia e Chirurgia Protesica e dei Reimpianti d'anca e di Ginocchio, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Maria Vittoria Marvi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Antonio Mazzotti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Cesare Faldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
- Ist Orthopedic Department, IRCCS Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Camilla Evangelisti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126, Bologna, Italy
| |
Collapse
|
25
|
Yang T, Cao T, Yang X, Wang G, Li Y. Elucidation of the key therapeutic targets and potential mechanisms of Andrographolide multi-targets against osteoarthritis via network pharmacological analysis and experimental validation. Gene 2024; 911:148351. [PMID: 38462021 DOI: 10.1016/j.gene.2024.148351] [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: 12/29/2023] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE Our purpose is to unveil Andrographolide's potential multi-target and multi-mechanism therapeutic effects in treating OA via systematic network pharmacological analysis and cell experimental validation. MATERIALS AND METHODS Initially, we gathered data from Andrographolide and OA-related databases to obtain information on Andrographolide's biological properties and the targets linked with OA. We developed a bioinformatic network about Andrographolide and OA, whereby we analyzed the network to identify potential therapeutic targets and mechanisms of action of Andrographolide. Subsequently, we used molecular docking to analyze the binding sites of Andrographolide to the target proteins. At the same time, SDF-1 was used to construct an OA cell model to verify the therapeutic effect of Andrographolide on OA and its effect on target proteins. RESULTS Our experimental results show that Andrographolide has excellent pharmaceutical properties, by Lipinski's rules for drugs, suggesting that this compound can be considered to have a high therapeutic potential in drug development. 233 targets were preliminarily investigated, the mechanisms through which Andrographolide targets OA primarily involve the TNF signaling pathway, PI3K-AKT signaling pathway, IL-17 signaling pathway, and TLR signaling pathway. These mechanisms target OA by influencing immune and inflammatory responses in the joints, regulating apoptosis to prevent chondrocyte death. Finally, TNF-α, STAT3, TP53, IL-6, JUN, IL-1β, HIF-1α, TGF-β1, and AKT1 were identified as 9 key targets of Andrographolide anti-OA. In addition, our molecular docking analyzes with cell experimental validation further confirm the network pharmacology results. According to our molecular docking results, Andrographolide can bind to all the hub target proteins and has a good binding ability (binding energy < -5 kcal/mol), with the strongest binding affinity to AKT1 of -9.2 kcal/ mol. The results of cell experiments showed that Andrographolide treatment significantly increased the cell viability and the expression of COL2A1 and ACAN proteins. Moreover, 30 μM Andrographolide significantly reversed SDF-1-induced increases in the protein expression of TNF-α, STAT3, TP53, IL-6, JUN, IL-1β, HIF-1α, and TGF-β1, and decreases in the protein expression of AKT1. CONCLUSION This study provides a comprehensive understanding of the potential therapeutic targets and mechanisms of action of Andrographolide in OA treatment. Our findings suggest that Andrographolide is a promising candidate for drug development in the management of OA.
Collapse
Affiliation(s)
- Tengyun Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Tingting Cao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Xianguang Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
26
|
Jia S, Liang R, Chen J, Liao S, Lin J, Li W. Emerging technology has a brilliant future: the CRISPR-Cas system for senescence, inflammation, and cartilage repair in osteoarthritis. Cell Mol Biol Lett 2024; 29:64. [PMID: 38698311 PMCID: PMC11067114 DOI: 10.1186/s11658-024-00581-x] [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: 12/29/2023] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Osteoarthritis (OA), known as one of the most common types of aseptic inflammation of the musculoskeletal system, is characterized by chronic pain and whole-joint lesions. With cellular and molecular changes including senescence, inflammatory alterations, and subsequent cartilage defects, OA eventually leads to a series of adverse outcomes such as pain and disability. CRISPR-Cas-related technology has been proposed and explored as a gene therapy, offering potential gene-editing tools that are in the spotlight. Considering the genetic and multigene regulatory mechanisms of OA, we systematically review current studies on CRISPR-Cas technology for improving OA in terms of senescence, inflammation, and cartilage damage and summarize various strategies for delivering CRISPR products, hoping to provide a new perspective for the treatment of OA by taking advantage of CRISPR technology.
Collapse
Affiliation(s)
- Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Rongji Liang
- Shantou University Medical College, Shantou, 515041, China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Shuai Liao
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Wei Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
27
|
Wang Z, Shi W, Wu L, Xiao Y, Wang M, Zhang S, Chen Z, Yin G, Xie X, Bi S, Liu S, Kong W, Zhou J. TMF inhibits extracellular matrix degradation by regulating the C/EBPβ/ADAMTS5 signaling pathway in osteoarthritis. Biomed Pharmacother 2024; 174:116501. [PMID: 38554527 DOI: 10.1016/j.biopha.2024.116501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/01/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease, characterized by degenerative destruction of articular cartilage. Chondrocytes, the unique cell type in cartilage, mediate the metabolism of extracellular matrix (ECM), which is mainly constituted by aggrecan and type II collagen. A disintegrin and metalloproteinase with thrombospondin 5 (ADAMTS5) is an aggrecanase responsible for the degradation of aggrecan in OA cartilage. CCAAT/enhancer binding protein β (C/EBPβ), a transcription factor in the C/EBP family, has been reported to mediate the expression of ADAMTS5. Our previous study showed that 5,7,3',4'-tetramethoxyflavone (TMF) could activate the Sirt1/FOXO3a signaling in OA chondrocytes. However, whether TMF protected against ECM degradation by down-regulating C/EBPβ expression was unknown. In this study, we found that aggrecan expression was down-regulated, and ADAMTS5 expression was up-regulated. Knockdown of C/EBPβ could up-regulate aggrecan expression and down-regulate ADAMTS5 expression in IL-1β-treated C28/I2 cells. TMF could compromise the effects of C/EBPβ on OA chondrocytes by activating the Sirt1/FOXO3a signaling. Conclusively, TMF exhibited protective activity against ECM degradation by mediating the Sirt1/FOXO3a/C/EBPβ pathway in OA chondrocytes.
Collapse
Affiliation(s)
- Zeyu Wang
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China
| | - Weimei Shi
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Yaosheng Xiao
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Miaofei Wang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Sainan Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Shengrong Bi
- Department of Joint Surgery, 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
|
28
|
Chen KT, Yeh CT, Yadav VK, Pikatan NW, Fong IH, Lee WH, Chiu YS. Notopterol mitigates IL-1β-triggered pyroptosis by blocking NLRP3 inflammasome via the JAK2/NF-kB/hsa-miR-4282 route in osteoarthritis. Heliyon 2024; 10:e28094. [PMID: 38532994 PMCID: PMC10963379 DOI: 10.1016/j.heliyon.2024.e28094] [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: 09/23/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Objective Osteoarthritis (OA), the most prevalent form of arthritis, impacts approximately 10% of men and 18% of women aged above 60 years. Currently, a complete cure for OA remains elusive, making clinical management challenging. The traditional Chinese herb Notopterygium incisum, integral to the Juanbi pill for rheumatism, shows promise in safeguarding chondrocytes through its strong anti-inflammatory effects. Methods To explore the protective effect of notopterol and miRNA (has-miR-4248) against inflammation, we simulated an inflammatory environment in chondrocytes cell lines C20A4 and C28/12, focusing on inflammasome formation and pyroptosis. Results Our finding indicates notopterol significantly reduced interleukin (IL)-18 and tumor necrosis factor (TNF)-alpha levels in inflamed cells, curtailed reactive oxygen species (ROS) production post-inflammation, and inhibited the JAK2/STAT3 signaling pathway, thus offering chondrocytes protection from inflammation. Importantly, notopterol also hindered inflammasome assembly and pyroptosis by blocking the NF-κB/NLRP3 pathway through hsa-miR-4282 modulation. In vivo experiments showed that notopterol treatment markedly decreased Osteoarthritis Research Society International (OARSI) scores in OA mice and boosted hsa-miR-4282 expression compared to control groups. Conclusions This study underscores notopterol's potential as a therapeutic agent in OA treatment, highlighting its capacity to shield cartilage from inflammation-induced damage, particularly by preventing pyroptosis.
Collapse
Affiliation(s)
- Ko-Ta Chen
- Department of Orthopedics, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung, 95092, Taiwan
| | - Vijesh Kumar Yadav
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Narpati Wesa Pikatan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Iat-Hang Fong
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Wei-Hwa Lee
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Yen-Shuo Chiu
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei, 11031, Taiwan
| |
Collapse
|
29
|
Yuan Z, Jiang D, Yang M, Tao J, Hu X, Yang X, Zeng Y. Emerging Roles of Macrophage Polarization in Osteoarthritis: Mechanisms and Therapeutic Strategies. Orthop Surg 2024; 16:532-550. [PMID: 38296798 PMCID: PMC10925521 DOI: 10.1111/os.13993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 02/02/2024] Open
Abstract
Osteoarthritis (OA) is the most common chronic degenerative joint disease in middle-aged and elderly people, characterized by joint pain and dysfunction. Macrophages are key players in OA pathology, and their activation state has been studied extensively. Various studies have suggested that macrophages might respond to stimuli in their microenvironment by changing their phenotypes to pro-inflammatory or anti-inflammatory phenotypes, which is called macrophage polarization. Macrophages accumulate and become polarized (M1 or M2) in many tissues, such as synovium, adipose tissue, bone marrow, and bone mesenchymal tissues in joints, while resident macrophages as well as other stromal cells, including fibroblasts, chondrocytes, and osteoblasts, form the joint and function as an integrated unit. In this study, we focus exclusively on synovial macrophages, adipose tissue macrophages, and osteoclasts, to investigate their roles in the development of OA. We review recent key findings related to macrophage polarization and OA, including pathogenesis, molecular pathways, and therapeutics. We summarize several signaling pathways in macrophage reprogramming related to OA, including NF-κB, MAPK, TGF-β, JAK/STAT, PI3K/Akt/mTOR, and NLRP3. Of note, despite the increasing availability of treatments for osteoarthritis, like intra-articular injections, surgery, and cellular therapy, the demand for more effective clinical therapies has remained steady. Therefore, we also describe the current prospective therapeutic methods that deem macrophage polarization to be a therapeutic target, including physical stimulus, chemical compounds, and biological molecules, to enhance cartilage repair and alleviate the progression of OA.
Collapse
Affiliation(s)
- Zimu Yuan
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Decheng Jiang
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Mengzhu Yang
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Jie Tao
- West China Medical SchoolSichuan UniversityChengduChina
- West China HospitalSichuan UniversityChengduChina
| | - Xin Hu
- Orthopedic Research Institute, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengduChina
| | - Xiao Yang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduChina
| | - Yi Zeng
- Orthopedic Research Institute, Department of OrthopedicsWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
30
|
Zhao Y, Mu Y, Zou Y, He Z, Lu T, Wang X, Li W, Gao B. Conjoint research of WGCNA, single-cell transcriptome and structural biology reveals the potential targets of IDD development and treatment and JAK3 involvement. Aging (Albany NY) 2023; 15:14764-14790. [PMID: 38095643 PMCID: PMC10781489 DOI: 10.18632/aging.205289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/02/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVES This study conducted integrated analysis of bulk RNA sequencing, single-cell RNA sequencing and Weighted Gene Co-expression Network Analysis (WGCNA), to comprehensively decode the most essential genes of intervertebral disc degeneration (IDD); then mainly focused on the JAK3 macromolecule to identify natural compounds to provide more candidate drug options in alleviating IDD. METHODS In the first part, we performed single-cell transcriptome analysis and WGCNA workflow to delineate the most pivotal genes of IDD. Then series of structural biology approaches and high-throughput virtual screening techniques were performed to discover potential compounds targeting JAK-STAT signaling pathway, such as Libdock, ADMET, precise molecular docking algorithm and in-vivo drug stability assessment. RESULTS Totally 4 hub genes were determined in the development of IDD, namely VEGFA, MMP3, TNFSF11, and TIMP3, respectively. Then, 3 novel natural materials, ZINC000014952116, ZINC000003938642 and ZINC000072131515, were determined as potential compounds, with less toxicities and moderate ADME characteristics. In-vivo drug stability assessment suggested that these drugs could interact with JAK3, and their ligand-JAK3 complexes maintained the homeostasis in-vivo, which acted as regulatory role to JAK3 protein. Among them, ZINC000072131515, also known as Menaquinone, demonstrated significant protective roles to alleviate the progression of IDD in vitro, which proved the nutritional therapy in alleviating IDD. CONCLUSIONS This study reported the essential genes in the development of IDD, and also the roles of Menaquinone to ameliorate IDD through inhibiting JAK3 protein. This study also provided more options and resources on JAK3 targeted screening, which may further expand the drug resources in the pharmaceutical market.
Collapse
Affiliation(s)
- Yingjing Zhao
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yuxue Mu
- Aerospace Clinical Medical Center, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Yujia Zou
- Department of Cardiology, Xinhua Hospital affiliated to School of Medicine, Shanghai Jiaotong University, China
| | - Zhijian He
- Department of Sports Teaching and Research, Lanzhou University, Lanzhou, China
| | - Tianxing Lu
- Zonglian College, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinhui Wang
- Department of Oncology, The Fifth Affiliated Hospital of Xinxiang Medical College, Xin Xiang 453100, China
| | - Weihang Li
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
31
|
Riggs KC, Sankar U. Inflammatory mechanisms in post-traumatic osteoarthritis: a role for CaMKK2. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00031. [PMID: 37849987 PMCID: PMC10578519 DOI: 10.1097/in9.0000000000000031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is a multifactorial disease of the cartilage, synovium, and subchondral bone resulting from direct joint trauma and altered joint mechanics after traumatic injury. There are no current disease-modifying therapies for PTOA, and early surgical interventions focused on stabilizing the joint do not halt disease progression. Chronic pain and functional disability negatively affect the quality of life and take an economic toll on affected patients. While multiple mechanisms are at play in disease progression, joint inflammation is a key contributor. Impact-induced mitochondrial dysfunction and cell death or altered joint mechanics after trauma culminate in inflammatory cytokine release from synoviocytes and chondrocytes, cartilage catabolism, suppression of cartilage anabolism, synovitis, and subchondral bone disease, highlighting the complexity of the disease. Current understanding of the cellular and molecular mechanisms underlying the disease pathology has allowed for the investigation of a variety of therapeutic strategies that target unique apoptotic and/or inflammatory processes in the joint. This review provides a concise overview of the inflammatory and apoptotic mechanisms underlying PTOA pathogenesis and identifies potential therapeutic targets to mitigate disease progression. We highlight Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2), a serine/threonine protein kinase that was recently identified to play a role in murine and human osteoarthritis pathogenesis by coordinating chondrocyte inflammatory responses and apoptosis. Given its additional effects in regulating macrophage inflammatory signaling and bone remodeling, CaMKK2 emerges as a promising disease-modifying therapeutic target against PTOA.
Collapse
Affiliation(s)
- Keegan C. Riggs
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
32
|
Park H, Lee S, Lee J, Moon H, Ro SW. Exploring the JAK/STAT Signaling Pathway in Hepatocellular Carcinoma: Unraveling Signaling Complexity and Therapeutic Implications. Int J Mol Sci 2023; 24:13764. [PMID: 37762066 PMCID: PMC10531214 DOI: 10.3390/ijms241813764] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatocellular Carcinoma (HCC) continues to pose a substantial global health challenge due to its high incidence and limited therapeutic options. In recent years, the Janus Kinase (JAK) and Signal Transducer and Activator of Transcription (STAT) pathway has emerged as a critical signaling cascade in HCC pathogenesis. The review commences with an overview of the JAK/STAT pathway, delving into the dynamic interplay between the JAK/STAT pathway and its numerous upstream activators, such as cytokines and growth factors enriched in pathogenic livers afflicted with chronic inflammation and cirrhosis. This paper also elucidates how the persistent activation of JAK/STAT signaling leads to diverse oncogenic processes during hepatocarcinogenesis, including uncontrolled cell proliferation, evasion of apoptosis, and immune escape. In the context of therapeutic implications, this review summarizes recent advancements in targeting the JAK/STAT pathway for HCC treatment. Preclinical and clinical studies investigating inhibitors and modulators of JAK/STAT signaling are discussed, highlighting their potential in suppressing the deadly disease. The insights presented herein underscore the necessity for continued research into targeting the JAK/STAT signaling pathway as a promising avenue for HCC therapy.
Collapse
Affiliation(s)
| | | | | | | | - Simon Weonsang Ro
- Department of Genetics and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si 17104, Republic of Korea; (H.P.); (S.L.); (J.L.); (H.M.)
| |
Collapse
|
33
|
Vignon C, Hilpert M, Toupet K, Goubaud A, Noël D, de Kalbermatten M, Hénon P, Jorgensen C, Barbero A, Garitaonandia I. Evaluation of expanded peripheral blood derived CD34+ cells for the treatment of moderate knee osteoarthritis. Front Bioeng Biotechnol 2023; 11:1150522. [PMID: 37288358 PMCID: PMC10242004 DOI: 10.3389/fbioe.2023.1150522] [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: 01/24/2023] [Accepted: 04/04/2023] [Indexed: 06/09/2023] Open
Abstract
Knee osteoarthritis (OA) is a degenerative joint disease of the knee that results from the progressive loss of articular cartilage. It is most common in the elderly and affects millions of people worldwide, leading to a continuous increase in the number of total knee replacement surgeries. These surgeries improve the patient's physical mobility, but can lead to late infection, loosening of the prosthesis, and persistent pain. We would like to investigate if cell-based therapies can avoid or delay such surgeries in patients with moderate OA by injecting expanded autologous peripheral blood derived CD34+ cells (ProtheraCytes®) into the articular joint. In this study we evaluated the survival of ProtheraCytes® when exposed to synovial fluid and their performance in vitro with a model consisting of their co-culture with human OA chondrocytes in separate layers of Transwells and in vivo with a murine model of OA. Here we show that ProtheraCytes® maintain high viability (>95%) when exposed for up to 96 hours to synovial fluid from OA patients. Additionally, when co-cultured with OA chondrocytes, ProtheraCytes® can modulate the expression of some chondrogenic (collagen II and Sox9) and inflammatory/degrading (IL1β, TNF, and MMP-13) markers at gene or protein levels. Finally, ProtheraCytes® survive after injection into the knee of a collagenase-induced osteoarthritis mouse model, engrafting mainly in the synovial membrane, probably due to the fact that ProtheraCytes® express CD44, a receptor of hyaluronic acid, which is abundantly present in the synovial membrane. This report provides preliminary evidence of the therapeutic potential of CD34+ cells on OA chondrocytes in vitro and their survival after in vivo implantation in the knee of mice and merits further investigation in future preclinical studies in OA models.
Collapse
Affiliation(s)
| | - Morgane Hilpert
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Karine Toupet
- Institute of Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, Montpellier, France
| | | | - Danièle Noël
- Institute of Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, Montpellier, France
| | | | | | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, Montpellier, France
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | | |
Collapse
|
34
|
Muhammad Firdaus FI, Nashihah AK, Mohd Fauzi MB, Manira M, Aminuddin S, Lokanathan Y. Application of Conditioned Medium for In Vitro Modeling and Repair of Respiratory Tissue. APPLIED SCIENCES 2023; 13:5862. [DOI: 10.3390/app13105862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background: The idea of exploring respiratory therapy in vitro predominantly guided by cell-secreted substances has gained ground in recent years. A conditioned medium (CM) consists of protein milieu that contains a diverse spectrum of cytokines, chemokines, angiogenic agents, and growth factors. This review evaluated the efficacy of using CM collected in an in vitro respiratory epithelial model. Methods: Twenty-six papers were included in this review: twenty-one cellular response studies on respiratory secretome application and five studies involving animal research. Results: The CM produced by differentiated cells from respiratory and non-respiratory systems, such as mesenchymal stem cells (MSC), exhibited the similar overall effect of improving proliferation and regeneration. Not only could differentiated cells from respiratory tissues increase proliferation, migration, and attachment, but the CM was also able to protect the respiratory epithelium against cytotoxicity. Most non-respiratory tissue CM was used as a treatment model to determine the effects of the therapy, while only one study used particle-based CM and reported decreased epithelial cell tight junctions, which harmed the epithelial barrier. Conclusion: As it resolves the challenges related to cell development and wound healing while simultaneously generally reducing the danger of immunological compatibility and tumorigenicity, CM might be a potential regenerative therapy in numerous respiratory illnesses. However, additional research is required to justify using CM in respiratory epithelium clinical practice.
Collapse
Affiliation(s)
- Fairuz Izan Muhammad Firdaus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ab. Karim Nashihah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh. Busra Mohd Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Maarof Manira
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Saim Aminuddin
- Graduate School of Medicine, KPJ Healthcare University College, Kota Seriemas, Nilai 71800, Malaysia
- KPJ Ampang Puteri Specialist Hospital, Ampang 68000, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
35
|
Liu S, Pan Y, Li T, Zou M, Liu W, Li Q, Wan H, Peng J, Hao L. The Role of Regulated Programmed Cell Death in Osteoarthritis: From Pathogenesis to Therapy. Int J Mol Sci 2023; 24:5364. [PMID: 36982438 PMCID: PMC10049357 DOI: 10.3390/ijms24065364] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Osteoarthritis (OA) is a worldwide chronic disease that can cause severe inflammation to damage the surrounding tissue and cartilage. There are many different factors that can lead to osteoarthritis, but abnormally progressed programmed cell death is one of the most important risk factors that can induce osteoarthritis. Prior studies have demonstrated that programmed cell death, including apoptosis, pyroptosis, necroptosis, ferroptosis, autophagy, and cuproptosis, has a great connection with osteoarthritis. In this paper, we review the role of different types of programmed cell death in the generation and development of OA and how the different signal pathways modulate the different cell death to regulate the development of OA. Additionally, this review provides new insights into the radical treatment of osteoarthritis rather than conservative treatment, such as anti-inflammation drugs or surgical operation.
Collapse
Affiliation(s)
- Suqing Liu
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Marry College, Nanchang University, Nanchang 330006, China
| | - Yurong Pan
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Marry College, Nanchang University, Nanchang 330006, China
| | - Ting Li
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Mi Zou
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wenji Liu
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qingqing Li
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Huan Wan
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Jie Peng
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liang Hao
- Department of Orthopedics, Second Affifiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|