1
|
Qian C, Wang Y, Yuan Q, Guo Y, Wang Y. Insights into the itaconate family: Immunomodulatory mechanisms and therapeutic potentials. Eur J Pharmacol 2025; 997:177542. [PMID: 40147573 DOI: 10.1016/j.ejphar.2025.177542] [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: 11/09/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
The itaconate family, comprising itaconate derivatives, endogenous isomers, and other related compounds, has demonstrated substantial immunoregulatory properties. These compounds exhibit significant therapeutic potential in various disease models by modulating metabolic pathways, signal transduction cascades, and post-translational modifications. In this review, we delineate the structural characteristics and biological functions of the members of the itaconate family and elucidate their immunomodulatory mechanisms. Additionally, we summarize the immunomodulatory effects of the itaconate family across various disease categories, including cardiovascular, liver, respiratory, bone and cartilage, neurological, and autoimmune diseases. This review aims to deepen our understanding of the itaconate family and its potential applications, providing new perspectives and therapeutic strategies for inflammatory disorders and autoimmune diseases.
Collapse
Affiliation(s)
- Chunlin Qian
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yueying Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuchen Guo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Xie Y, Cheng Q, Xu ML, Xue J, Wu H, Du Y. Itaconate: A Potential Therapeutic Strategy for Autoimmune Disease. Scand J Immunol 2025; 101:e70026. [PMID: 40289463 DOI: 10.1111/sji.70026] [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: 03/04/2025] [Revised: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025]
Abstract
Itaconate is a metabolite of the Krebs cycle, and endogenous itaconate is driven by a variety of innate signals that inhibit the production of inflammatory cytokines. The key mechanism of action of itaconate was initially found to be the competitive inhibition of succinate dehydrogenase (SDH), which inhibits the production of inflammatory factors, as well as its antioxidant effects. With increasing research, it was discovered that it modifies cysteine residues of related proteins through the Michael addition, such as modifying the Kelch-like ECH-associated protein 1 (KEAP1) protein and activating the nuclear factor erythroid 2-related factor 2 (NRF2) signalling pathway, as well as glycolytic enzymes and cellular pathway-associated factors that attenuate inflammatory responses and oxidative stress. It also acts on a variety of immune cells, affecting their function and activity, and has been increasingly shown to play a therapeutic role in a variety of inflammatory and autoimmune diseases through a combination of these mechanisms. In conclusion, there has been a great breakthrough in the research of itaconate, from the initial industrial application to the redefinition of the biological functions of itaconate. However, with the deepening of the research, we also found that there are more questions: the mechanism of action of itaconate, more functions of itaconate, clinical application of itaconate, and the use of itaconate still needs to be solved.
Collapse
Affiliation(s)
- Yifan Xie
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Clinic Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Cheng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Li Xu
- Department of Nephrology, The Third Affiliate Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Xue
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Du
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Henry ÓC, O'Neill LAJ. Metabolic Reprogramming in Stromal and Immune Cells in Rheumatoid Arthritis and Osteoarthritis: Therapeutic Possibilities. Eur J Immunol 2025; 55:e202451381. [PMID: 40170391 PMCID: PMC11962241 DOI: 10.1002/eji.202451381] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/03/2025]
Abstract
Metabolic reprogramming of stromal cells, including fibroblast-like synoviocytes (FLS) and chondrocytes, as well as osteoclasts (OCs), are involved in the inflammatory and degenerative processes underlying rheumatoid arthritis (RA) and osteoarthritis (OA). In RA, FLS exhibit mTOR activation, enhanced glycolysis and reduced oxidative phosphorylation, fuelling inflammation, angiogenesis, and cartilage degradation. In OA, chondrocytes undergo metabolic rewiring, characterised by mTOR and NF-κB activation, mitochondrial dysfunction, and increased glycolysis, which promotes matrix metalloproteinase production, extracellular matrix (ECM) degradation, and angiogenesis. Macrophage-derived immunometabolites, including succinate and itaconate further modulate stromal cell function, acting as signalling molecules that modulate inflammatory and catabolic processes. Succinate promotes inflammation whilst itaconate is anti-inflammatory, suppressing inflammatory joint disease in models. Itaconate deficiency also correlates inversely with disease severity in RA in humans. Emerging evidence highlights the potential of targeting metabolic processes as promising therapeutic strategies for connective tissue disorders.
Collapse
Affiliation(s)
- Órlaith C. Henry
- Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | | |
Collapse
|
4
|
Gao X, Tang M, Li J, Ma J, Liu Z, Liu W. Activation of Nrf2 pathway by 4-Octyl itaconate enhances donor lung function in cold preservation settings. Respir Res 2025; 26:69. [PMID: 40016745 PMCID: PMC11869626 DOI: 10.1186/s12931-025-03151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Lung transplantation is the primary treatment for end-stage lung diseases. However, ischemia-reperfusion injury (IRI) significantly impacts transplant outcomes. 4-Octyl itaconate (4-OI) has shown potential in mitigating organ IRI, although its effects in lung transplantation require further exploration. METHODS BEAS-2B cells were used to model transplantation, assessing the effects of 4-OI through viability, apoptosis, and ROS assays. qRT-PCR analyzed cytokine transcription post-cold ischemia/reperfusion (CI/R). RNA sequencing and Gene Ontology analysis elucidated 4-OI's mechanisms of action, confirmed by Western blotting. ALI-airway and lung transplantation organoid models evaluated improvements in bronchial epithelial morphology and function due to 4-OI. ELISA measured IL-6 and IL-8 levels. Rat models of extended cold preservation and non-heart-beating transplantation assessed 4-OI's impact on lung function, injury, and inflammation. RESULTS Our findings indicate that 4-OI (100 µM) during cold preservation effectively maintained cell viability, decreased apoptosis, and reduced ROS production in BEAS-2B cells under CI/R conditions. It also downregulated pro-inflammatory cytokine transcription, including IL1B, IL6, and TNF. Inhibition of Nrf2 partially reversed these protective effects. In cold preservation solutions, 4-OI upregulated Nrf2 target genes such as NQO1, HMOX1, and SLC7A11. In ALI airway models, 4-OI enhanced bronchial epithelial barrier integrity and ciliary beat function after CI/R. In rat models, 4-OI administration improved lung function and reduced pulmonary edema, tissue injury, apoptosis, and systemic inflammation following extended cold preservation or non-heart-beating lung transplantation. CONCLUSIONS Incorporating 4-OI into cold preservation solutions appears promising for alleviating CI/R-induced bronchial epithelial injury and enhancing lung transplant outcomes via Nrf2 pathway activation.
Collapse
Affiliation(s)
- Xinliang Gao
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jialin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jianzun Ma
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhengrui Liu
- Changchun Yifu Jilin Province Academician Workstation, Changchun, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
5
|
Sun Q, Nan XY, Wang H, Pan S, Ji G, Guo YF, Zhao YH, Li GC, Guo SS, Lin LF, Jin YJ, Zhang XL, Liu CC, Liu GB. Polydatin retards the progression of osteoarthritis by maintaining bone metabolicbalance and inhibiting macrophage polarization. Front Bioeng Biotechnol 2025; 12:1514483. [PMID: 39840130 PMCID: PMC11747576 DOI: 10.3389/fbioe.2024.1514483] [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: 10/21/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Background Polydatin (PD), also known as tiger cane glycoside, is a natural compound extracted from the Japanese knotweed plant, which is often referred to as white resveratrol. It exhibits anti-inflammatory, antioxidant, and anti-apoptotic effects in the treatment of various diseases. However, the potential molecular mechanisms of PD in osteoarthritis have not been clearly elucidated. Methods Anterior cruciate ligament transection (ACLT) surgery was performed to establish an osteoarthritis animal model. Female mice at the age of 12 weeks were intraperitoneally injected with different concentrations of PD (20 and 40 mg/kg). In vitro models were established by isolating mouse articular chondrocytes, which were subsequently treated with lipopolysaccharide or IL-1β for 24 h for subsequent experiments. In addition, different concentrations of PD were administered for 12 h. Morphological changes were observed by toluidine blue staining, joint bone metabolism changes were observed by tartrate-resistant acid phosphatase staining, immunohistochemistry was used to observe the expression levels of inflammatory factors and extracellular matrix. MicroCT analysis was conducted to assess changes in the microstructure of subchondral bone trabeculae, and Western blot was performed to measure the expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway and markers of M1 polarization in macrophages. Results PD significantly delays the progression of osteoarthritis induced by ACLT, effectively inhibits IL-1β-induced joint inflammation, bone metabolic remodeling and extracellular matrix degradation. In addition, paeoniflorin markedly suppresses the transmission of the NF-κB signaling pathway and reverses M1 polarization in macrophages induced by IL-1β. Conclusion Taken together, PD might be a potential therapeutic agent for the prevention and treatment of osteoarthritis.
Collapse
Affiliation(s)
- Qi Sun
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xin-Yu Nan
- Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hui Wang
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuo Pan
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Gang Ji
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ya-Feng Guo
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ya-Heng Zhao
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Gao-Cen Li
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shao-Shi Guo
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lu-Feng Lin
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu-Jie Jin
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xue Li Zhang
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chang-Cheng Liu
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Guo-Bin Liu
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
6
|
Tang YZ, Chen W, Xu BY, He G, Fan XC, Tang KL. 4-Octyl itaconate inhibits synovitis in the mouse model of post-traumatic osteoarthritis and alleviates pain. Chin J Traumatol 2025; 28:50-61. [PMID: 39578202 PMCID: PMC11840323 DOI: 10.1016/j.cjtee.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/21/2024] [Accepted: 10/08/2024] [Indexed: 11/24/2024] Open
Abstract
PURPOSE To investigate the pathological changes of the synovium in mice with post-traumatic osteoarthritis (PTOA) treated with 4-octyl itaconate (4-OI) and evaluate the therapeutic effects of 4-OI. METHODS In the phenotypic validation experiment, the mice were randomly divided into 3 groups: wild-type (WT) group, sham group, and destabilization of the medial meniscus (DMM) group. Through MRI, micro-CT, and histological analysis, it was determined that the DMM surgery induced a mouse PTOA model with significant signs of synovitis. At 12 weeks post-DMM surgery, synovial tissues from the DMM group and WT group mice were collected for ribonucleic acid sequencing analysis. In the 4-OI treatment experiment, mice were randomly divided into the sham group, DMM group, DMM + 4-OI (50 mg/kg) group, and DMM + 4-OI (100 mg/kg) group. Von Frey tests and open field tests were conducted at intervals during the 12 weeks following the DMM surgery. After 12 weeks of surgery, the efficacy of 4-OI treatment on PTOA in mice was evaluated using MRI, micro-CT, histological analysis, and quantitative real-time polymerase chain reaction. Finally, we utilized network pharmacology analysis to predict the mechanism of 4-OI in treating PTOA synovitis and conducted preliminary validation. Statistical analysis was performed using one-way ANOVA and the Kruskal-Wallis test. Difference was considered statistically significant at p < 0.05. RESULTS The DMM surgery effectively induced a PTOA mouse model, which displayed significant symptoms of synovitis. These symptoms included a notable increase in both the number of calcified tissues and osteophytes (p < 0.001), an enlargement of the calcified meniscus and synovial tissue volume (p < 0.001), and thickening of the synovial lining layer attributable to M1 macrophage accumulation (p = 0.035). Additionally, we observed elevated histological scores for synovitis (p < 0.001). Treatment with 4-OI inhibited the thickening of M1 macrophages in the synovial lining layer of PTOA mice (p < 0.001) and reduced fibrosis in the synovial stroma (p = 0.004). Furthermore, it reduced the histological scores of knee synovitis in PTOA mice (p = 0.006) and improved the inflammatory microenvironment associated with synovitis. Consequently, this treatment alleviated pain in PTOA mice (p < 0.001) and reduced spontaneous activity (p = 0.003). Bioinformatics and network pharmacology analyses indicated that 4-OI may exert its therapeutic effects by inhibiting the differentiation of synovial Th17 cells. Specifically, compared to the lipopolysaccharide stimulation group, 4-OI reduced the levels of positive regulatory factors of Th17 cell differentiation (IL-1: p < 0.001, IL-6: p < 0.001), key effector molecules (IL-17A: p < 0.001, IL-17F: p = 0.004), and downstream effector molecules in the IL-17 signaling pathway (CCL2: p < 0.001, MMP13: p < 0.001). CONCLUSION 4-OI is effective in inhibiting synovitis in PTOA, thereby alleviating the associated painful symptoms.
Collapse
Affiliation(s)
- Yu-Zhen Tang
- Department of Orthopaedics/Sport Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400000, China
| | - Wan Chen
- Department of Orthopaedics/Sport Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400000, China.
| | - Bao-Yun Xu
- Department of Orthopaedics/Sport Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400000, China
| | - Gang He
- Department of Orthopaedics/Sport Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400000, China
| | - Xiu-Cheng Fan
- Department of Orthopaedics/Sport Medicine Center, First Affiliated Hospital of Army Medical University, Chongqing, 400000, China
| | - Kang-Lai Tang
- Department of Training Injury Prevention, Army Medical University, Chongqing, 400000, China.
| |
Collapse
|
7
|
Li R, Sun K. Regulation of chondrocyte apoptosis in osteoarthritis by endoplasmic reticulum stress. Cell Stress Chaperones 2024; 29:750-763. [PMID: 39515603 PMCID: PMC11626768 DOI: 10.1016/j.cstres.2024.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA), a common degenerative joint disease, is characterized by the apoptosis of chondrocytes as a primary pathophysiological change, with endoplasmic reticulum stress (ERS) playing a crucial role. It has been demonstrated that an imbalance in endoplasmic reticulum (ER) homeostasis can lead to ERS, activating three cellular adaptive response pathways through the unfolded protein response to restore ER homeostasis. Mild ERS exerts a protective effect on cells, while prolonged ERS that disrupts the self-regulatory balance of the ER activates apoptotic signaling pathways, leading to chondrocyte apoptosis and hastening OA progression. Hence, controlling the ERS signaling pathway and its apoptotic factors has become a critical focus for preventing and treating OA. This review aims to elucidate the key mechanisms of ERS pathway-induced apoptosis, associated targets, and regulatory pathways, offering valuable insights to enhance the mechanistic understanding of OA. It also reviews the mechanisms studied for ERS-related drugs or compounds for the treatment of OA.
Collapse
Affiliation(s)
- Renzhong Li
- Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu Province, China; The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, China
| | - Kui Sun
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, China; Anhui Acupuncture Hospital, Hefei, Anhui Province, China.
| |
Collapse
|
8
|
Li L, Li J, Li JJ, Zhou H, Zhu XW, Zhang PH, Huang B, Zhao WT, Zhao XF, Chen ES. Chondrocyte autophagy mechanism and therapeutic prospects in osteoarthritis. Front Cell Dev Biol 2024; 12:1472613. [PMID: 39507422 PMCID: PMC11537998 DOI: 10.3389/fcell.2024.1472613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Osteoarthritis (OA) is the most common type of arthritis characterized by progressive cartilage degradation, with its pathogenesis closely related to chondrocyte autophagy. Chondrocytes are the only cells in articular cartilage, and the function of chondrocytes plays a vital role in maintaining articular cartilage homeostasis. Autophagy, an intracellular degradation system that regulates energy metabolism in cells, plays an incredibly important role in OA. During the early stages of OA, autophagy is enhanced in chondrocytes, acting as an adaptive mechanism to protect them from various environmental changes. However, with the progress of OA, chondrocyte autophagy gradually decreases, leading to the accumulation of damaged organelles and macromolecules within the cell, prompting chondrocyte apoptosis. Numerous studies have shown that cartilage degradation is influenced by the senescence and apoptosis of chondrocytes, which are associated with reduced autophagy. The relationship between autophagy, senescence, and apoptosis is complex. While autophagy is generally believed to inhibit cellular senescence and apoptosis to promote cell survival, recent studies have shown that some proteins are degraded by selective autophagy, leading to the secretion of the senescence-associated secretory phenotype (SASP) or increased SA-β-Gal activity in senescent cells within the damaged region of human OA cartilage. Autophagy activation may lead to different outcomes depending on the timing, duration, or type of its activation. Thus, our study explored the complex relationship between chondrocyte autophagy and OA, as well as the related regulatory molecules and signaling pathways, providing new insights for the future development of safe and effective drugs targeting chondrocyte autophagy to improve OA.
Collapse
Affiliation(s)
- Lan Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian-Jiang Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Huan Zhou
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xing-Wang Zhu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Ping-Heng Zhang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Huang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen-Ting Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao-Feng Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - En-Sheng Chen
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Zhang Y, Zhou Y. Advances in targeted therapies for age-related osteoarthritis: A comprehensive review of current research. Biomed Pharmacother 2024; 179:117314. [PMID: 39167845 DOI: 10.1016/j.biopha.2024.117314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that disproportionately impacts the elderly population on a global scale. As aging is a significant risk factor for OA, there is a growing urgency to develop specific therapies that target the underlying mechanisms of aging associated with this condition. This summary seeks to offer a thorough introduction of ongoing research efforts aimed at developing therapies to combat senescence in the context of OA. Cellular senescence plays a pivotal role in both the deterioration of cartilage integrity and the perpetuation of chronic inflammation and tissue remodeling. Consequently, targeting SnCs has emerged as a promising therapeutic approach to alleviate symptoms and hinder the progression of OA. This review examines a range of approaches, including senolytic drugs targeting SnCs, senomorphics that modulate the senescence-associated secretory phenotype (SASP), and interventions that enhance immune system clearance of SnCs. Novel methodologies, such as utilizing novel materials for exosome delivery and administering anti-aging medications with precision, offer promising avenues for the precise treatment of OA. Accumulating evidence underscores the potential of targeting senescence in OA management, potentially facilitating the development of more effective and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Yantao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuan 430060, China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuan 430060, China.
| |
Collapse
|
10
|
Pan X, Kong X, Feng Z, Jin Z, Wang M, Lu H, Chen G. 4-Octyl itaconate protects chondrocytes against IL-1β-induced oxidative stress and ferroptosis by inhibiting GPX4 methylation in osteoarthritis. Int Immunopharmacol 2024; 137:112531. [PMID: 38906009 DOI: 10.1016/j.intimp.2024.112531] [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: 02/12/2024] [Revised: 06/02/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
The role of oxidative stress and ferroptosis in osteoarthritis (OA) pathogenesis is increasingly recognized. Notably, 4-octyl Itaconate (OI) has been documented to counteract oxidative stress and inflammatory responses, highlighting its therapeutic potential in OA. This study explored the effects of OI on GPX4 methylation, oxidative stress, and ferroptosis in chondrocytes affected by OA. Our results demonstrated that OI mitigated IL-1β-induced chondrocyte degeneration in a dose-dependent manner. It also suppressed reactive oxygen species (ROS) production and sustained GPX4 expression, thereby attenuating the degenerative impact of IL-1β and Erastin on chondrocytes by curtailing ferroptosis. Moreover, we observed that blocking GPX4 methylation could alleviate IL-1β-induced degeneration, oxidative stress, and ferroptosis in chondrocytes. The regulatory mechanism of OI on GPX4 expression in chondrocytes involved the inhibition of GPX4 methylation. In a mouse model of OA, OI's protective effects against OA were comparable to those of Ferrostatin-1. Thus, OI reduced chondrocyte degeneration, oxidative stress, and ferroptosis by inhibiting GPX4 methylation, offering a novel mechanistic insight into its therapeutic application in OA.
Collapse
Affiliation(s)
- Xuekang Pan
- Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Xiangjia Kong
- Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Zhenhua Feng
- Sir Run Run Shaw Hospital, Hangzhou 310000, China
| | - Zheyuan Jin
- Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Mige Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China.
| | - Huigen Lu
- Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China.
| | - Gang Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, China.
| |
Collapse
|
11
|
Wu QJ, Li Q, Yang P, Du L. Itaconate to treat acute lung injury: recent advances and insights from preclinical models. Am J Transl Res 2024; 16:3480-3497. [PMID: 39262751 PMCID: PMC11384376 DOI: 10.62347/nuin2087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/22/2024] [Indexed: 09/13/2024]
Abstract
Acute lung injury (ALI) is defined as the acute onset of diffuse bilateral pulmonary infiltration, leading to PaO2/FiO2 ≤ 300 mmHg without clinical evidence of left atrial hypertension. Acute respiratory distress syndrome (ARDS) involves more severe hypoxemia (PaO2/FiO2 ≤ 200 mmHg). Treatment of ALI and ARDS has received renewed attention as the incidence of ALI caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has increased. Itaconate and its derivatives have shown therapeutic potential against ALI. This review provides an in-depth summary of the mechanistic research of itaconate in the field of acute lung injury, including inducing autophagy, preventing ferroptosis and pyroptosis, shifting macrophage polarization to an anti-inflammatory M2 phenotype, inhibiting neutrophil activation, regulating epigenetic modifications, and repressing aerobic glycolysis. These compounds merit further consideration in clinical trials. We anticipate that the clinical translation of itaconate-based drugs can be accelerated.
Collapse
Affiliation(s)
- Qin Juan Wu
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
- Department of Anesthesiology, Chengdu Second People's Hospital Chengdu 610000, Sichuan, China
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
| | - Ping Yang
- Department of Anesthesiology, Chongqing University Three Gorges Hospital Chongqing 404100, China
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
| | - Lei Du
- Department of Anesthesiology, West China Hospital of Sichuan University Chengdu 610041, Sichuan, China
| |
Collapse
|
12
|
Kong X, Xu L, Mou Z, Lyu W, Shan K, Wang L, Liu F, Rong F, Li J, Wei P. The anti-inflammatory effects of itaconate and its derivatives in neurological disorders. Cytokine Growth Factor Rev 2024; 78:37-49. [PMID: 38981775 DOI: 10.1016/j.cytogfr.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Almost 16 % of the global population is affected by neurological disorders, including neurodegenerative and cerebral neuroimmune diseases, triggered by acute or chronic inflammation. Neuroinflammation is recognized as a common pathogenic mechanism in a wide array of neurological conditions including Alzheimer's disease, Parkinson's disease, postoperative cognitive dysfunction, stroke, traumatic brain injury, and multiple sclerosis. Inflammatory process in the central nervous system (CNS) can lead to neuronal damage and neuronal apoptosis, consequently exacerbating these diseases. Itaconate, an immunomodulatory metabolite from the tricarboxylic acid cycle, suppresses neuroinflammation and modulates the CNS immune response. Emerging human studies suggest that itaconate levels in plasma and cerebrospinal fluid may serve as biomarkers associated with inflammatory responses in neurological disorders. Preclinical studies have shown that itaconate and its highly cell-permeable derivatives are promising candidates for preventing and treating neuroinflammation-related neurological disorders. The underlying mechanism may involve the regulation of immune cells in the CNS and neuroinflammation-related signaling pathways and molecules including Nrf2/KEAP1 signaling pathway, reactive oxygen species, and NLRP3 inflammasome. Here, we introduce the metabolism and function of itaconate and the synthesis and development of its derivatives. We summarize the potential impact and therapeutic potential of itaconate and its derivatives on brain immune cells and the associated signaling pathways and molecules, based on preclinical evidence via various neurological disorder models. We also discuss the challenges and potential solutions for clinical translation to promote further research on itaconate and its derivatives for neuroinflammation-related neurological disorders.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Lin Xu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Zheng Mou
- Department of Pharmacy, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Wenyuan Lyu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Kaiyue Shan
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Longfei Wang
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Fanghao Liu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Fei Rong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Jianjun Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Penghui Wei
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China; Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
| |
Collapse
|
13
|
Xu T, Liu K, Fan J, Jia X, Guo X, Zhao X, Cao Y, Zhang H, Wang Q. Metformin mitigates osteoarthritis progression by modulating the PI3K/AKT/mTOR signaling pathway and enhancing chondrocyte autophagy. Open Life Sci 2024; 19:20220922. [PMID: 39091625 PMCID: PMC11292032 DOI: 10.1515/biol-2022-0922] [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: 04/09/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 08/04/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease characterized by overall joint tissue damage. Metformin (Met) has been shown to inhibit inflammatory reactions, though its potential protective mechanism on cartilage remains unclear. This study investigated Met's potential to protect cartilage in an OA rat model. Various morphological experiments were conducted to assess changes in cartilage tissue morphology before and after Met treatment. Protein and mRNA levels of cartilage-specific genes were measured using western blot, immunohistochemical staining, and RT-qPCR. Additionally, protein levels of autophagy-related and mTOR pathway-related proteins were measured. The results indicate an imbalance in the synthesis and degradation metabolism of chondrocytes, downregulation of cellular autophagy, and activation of the PI3K/Akt/mTOR pathway after surgery. However, treatment with Met could upregulate the expression of synthetic metabolic factors, indicating its contribution to cartilage repair. Furthermore, analysis of autophagy and pathway protein levels indicated that Met effectively attenuated autophagic damage to osteoarthritic cartilage cells and abnormal activation of the PI3K/Akt/mTOR pathway. In conclusion, Met can inhibit the abnormal activation of the PI3K/AKT/mTOR signaling pathway in cartilage tissue, promote the restoration of cartilage cell autophagic function, improve the balance of cartilage cell synthesis and degradation metabolism, and thus exert a protective effect on rat joint cartilage.
Collapse
Affiliation(s)
- Tianjie Xu
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063000, China
- Hebei Key Laboratory for Chronic Diseases, Tangshan, Hebei, 063000, China
| | - Kainan Liu
- Department of Basic Medicine, Xingtai Medical College, Xingtai, Hebei, 054000, China
| | - Jiaxin Fan
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063000, China
- Hebei Key Laboratory for Chronic Diseases, Tangshan, Hebei, 063000, China
| | - Xiang Jia
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063000, China
- Hebei Key Laboratory for Chronic Diseases, Tangshan, Hebei, 063000, China
| | - Xiaoling Guo
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063000, China
- Hebei Key Laboratory for Chronic Diseases, Tangshan, Hebei, 063000, China
| | - Xingwang Zhao
- Department of Orthopedics, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, 063000, China
| | - Yanhua Cao
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063000, China
| | - Hui Zhang
- Department of Joint Surgery 1, The Second Hospital of Tangshan, Tangshan, Hebei, 063000, China
| | - Qian Wang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei, 063000, China
- Hebei Key Laboratory for Chronic Diseases, Tangshan, Hebei, 063000, China
| |
Collapse
|
14
|
Lawrence M, Goyal A, Pathak S, Ganguly P. Cellular Senescence and Inflammaging in the Bone: Pathways, Genetics, Anti-Aging Strategies and Interventions. Int J Mol Sci 2024; 25:7411. [PMID: 39000517 PMCID: PMC11242738 DOI: 10.3390/ijms25137411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Advancing age is associated with several age-related diseases (ARDs), with musculoskeletal conditions impacting millions of elderly people worldwide. With orthopedic conditions contributing towards considerable number of patients, a deeper understanding of bone aging is the need of the hour. One of the underlying factors of bone aging is cellular senescence and its associated senescence associated secretory phenotype (SASP). SASP comprises of pro-inflammatory markers, cytokines and chemokines that arrest cell growth and development. The accumulation of SASP over several years leads to chronic low-grade inflammation with advancing age, also known as inflammaging. The pathways and molecular mechanisms focused on bone senescence and inflammaging are currently limited but are increasingly being explored. Most of the genes, pathways and mechanisms involved in senescence and inflammaging coincide with those associated with cancer and other ARDs like osteoarthritis (OA). Thus, exploring these pathways using techniques like sequencing, identifying these factors and combatting them with the most suitable approach are crucial for healthy aging and the early detection of ARDs. Several approaches can be used to aid regeneration and reduce senescence in the bone. These may be pharmacological, non-pharmacological and lifestyle interventions. With increasing evidence towards the intricate relationship between aging, senescence, inflammation and ARDs, these approaches may also be used as anti-aging strategies for the aging bone marrow (BM).
Collapse
Affiliation(s)
- Merin Lawrence
- School of Biological and Chemical Sciences, University of Galway, H91W2TY Galway, Ireland
| | - Abhishek Goyal
- RAS Life Science Solutions, Stresemannallee 61, 60596 Frankfurt, Germany
| | - Shelly Pathak
- Observational and Pragmatic Research Institute, 5 Coles Lane, Oakington, Cambridge CB24 3BA, UK
| | - Payal Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK
| |
Collapse
|
15
|
Li Y, Ding Z, Liu F, Li S, Huang W, Zhou S, Han Y, Liu L, Li Y, Yin Z. Luteolin regulating synthesis and catabolism of osteoarthritis chondrocytes via activating autophagy. Heliyon 2024; 10:e31028. [PMID: 38882274 PMCID: PMC11176761 DOI: 10.1016/j.heliyon.2024.e31028] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/18/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent bone and joint disease characterized by degeneration. The dysregulation between chondrocyte synthesis and breakdown is a key factor in OA development. Targeting the degenerative changes in cartilage tissue degradation could be a potential treatment approach for OA. Previous research has established a strong link between autophagy and the regulation of chondrocyte functions. Activating autophagy has shown promise in mitigating cartilage tissue degeneration. Currently, osteoarthritis treatment primarily focuses on symptom management, as there is no definitive medication to stop disease progression. Previous studies have demonstrated that luteolin, a flavonoid present in Chinese herbal medicine, can activate autophagy and reduce the expression of MMP1 and ADAMTS-5. This study utilized an in vitro osteoarthritis model with chondrocytes stimulated by IL-1β, treated with varying concentrations of luteolin. Treatment with luteolin notably increased the levels of synthesis factors Aggrecan and Collagen II, while decreasing the levels of decomposition factors MMP-1 and ADAMTS-5. Moreover, inhibition of autophagy by Chloroquine reversed the imbalances in chondrocyte activities induced by IL-1β. In an in vivo model of knee osteoarthritis induced by medial meniscal instability (DMM), luteolin was administered as a therapeutic regimen. After 12 weeks, knee cartilage tissues from mice were analyzed. Immunofluorescence and immunohistochemical staining revealed a decrease in P62 expression and an increase in Beclin-1 in the cartilage tissues. Additionally, cartilage wear in the knee joints of mice was alleviated by safranin O and fast green staining. Our study findings underscore the significant role of luteolin in effectively rebalancing chondrocyte activities disrupted by IL-1β. Our results strongly indicate that luteolin has the potential to be developed as a novel therapeutic agent for the treatment of osteoarthritis, offering promising prospects for future drug development.
Collapse
Affiliation(s)
- Yetian Li
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, #218 Ji Xi Road, Hefei, 230032, Anhui, China
| | - Zhenfei Ding
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, #287 Changhuai Road, Bengbu, 233000, Anhui, China
| | - Fuen Liu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, #218 Ji Xi Road, Hefei, 230032, Anhui, China
- Department of Emergency Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, #17 Lu Jiang Road, Hefei, 230001, Anhui, China
| | - Shuang Li
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, #218 Ji Xi Road, Hefei, 230032, Anhui, China
| | - Wei Huang
- Department of Orthopedics,The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, #17 Lu Jiang Road, Hefei, 230001, Anhui, China
| | - Shusheng Zhou
- Department of Emergency Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, #17 Lu Jiang Road, Hefei, 230001, Anhui, China
| | - Yongsheng Han
- Department of Emergency Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, #17 Lu Jiang Road, Hefei, 230001, Anhui, China
| | - Ling Liu
- Department of Emergency Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, #17 Lu Jiang Road, Hefei, 230001, Anhui, China
| | - Yan Li
- Department of Emergency Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, #17 Lu Jiang Road, Hefei, 230001, Anhui, China
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, #218 Ji Xi Road, Hefei, 230032, Anhui, China
| |
Collapse
|
16
|
Lee YT, Mohd Yunus MH, Yazid MD, Ugusman A. Unraveling the path to osteoarthritis management: targeting chondrocyte apoptosis for therapeutic intervention. Front Cell Dev Biol 2024; 12:1347126. [PMID: 38827524 PMCID: PMC11140145 DOI: 10.3389/fcell.2024.1347126] [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: 11/30/2023] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
Osteoarthritis (OA) is a chronic disease affecting joints and further causing disabilities. This disease affects around 240 million people worldwide. It is a multifactorial disease, and its etiology is difficult to determine. Although numerous therapeutic strategies are available, the therapies are aimed at reducing pain and improving patients' quality of life. Hence, there is an urgent need to develop disease-modifying drugs (DMOAD) that can reverse or halt OA progression. Apoptosis is a cell removal process that is important in maintaining homeostatic mechanisms in the development and sustaining cell population. The apoptosis of chondrocytes is believed to play an important role in OA progression due to poor chondrocytes self-repair abilities to maintain the extracellular matrix (ECM). Hence, targeting chondrocyte apoptosis can be one of the potential therapeutic strategies in OA management. There are various mediators and targets available to inhibit apoptosis such as autophagy, endoplasmic reticulum (ER) stress, oxidative stress, and inflammation. As such, this review highlights the importance and potential targets that can be aimed to reduce chondrocyte apoptosis.
Collapse
Affiliation(s)
- Yi Ting Lee
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Mohd Heikal Mohd Yunus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| |
Collapse
|
17
|
Liu L, Wang J, Liu L, Shi W, Gao H, Liu L. WITHDRAWN: The dysregulated autophagy in osteoarthritis: Revisiting molecular profile. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024:S0079-6107(24)00034-8. [PMID: 38531488 DOI: 10.1016/j.pbiomolbio.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/21/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.
Collapse
Affiliation(s)
- Liang Liu
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Jie Wang
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Lu Liu
- Department of Internal Medicine, Tianbao Central Health Hospital, Xintai City, Shandong Province, Shandong, Xintai, 271200, China
| | - Wenling Shi
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| | - Huajie Gao
- Operating Room of Qingdao University Affiliated Hospital, Qingdao, Pingdu, 266000, China
| | - Lun Liu
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Pingdu, 266000, China
| |
Collapse
|
18
|
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
|
19
|
Zhang L, Wang Y, Tian L, Li L, Chen Z, Ding C, Tian J, Song D, Yao S, Ren W. Thrombospondin-1-mediated crosstalk between autophagy and oxidative stress orchestrates repair of blast lung injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167026. [PMID: 38237742 DOI: 10.1016/j.bbadis.2024.167026] [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: 08/31/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
Coal mining carries inherent risks of catastrophic gas explosions capable of inflicting severe lung injury. Using complementary in vivo and in vitro models, we explored mechanisms underlying alveolar epithelial damage and repair following a gas explosion in this study. In a rat model, the gas explosion was demonstrated to trigger inflammation and injury within the alveolar epithelium. The following scRNA-sequencing revealed that alveolar epithelial cells exhibited the most profound transcriptomic changes after gas explosion compared to other pulmonary cell types. In the L2 alveolar epithelial cells, the blast was found to cause autophagic flux by inducing autophagosome formation, LC3 lipidation, and p62 degradation. Transcriptomic profiling of the L2 cells identified PI3K-Akt and p53 pathways as critical modulators governing autophagic and oxidative stress responses to blast damage. Notably, Thrombospondin-1 (Thbs1) was determined for the first time as a pivotal node interconnecting these two pathways. The findings of this study illuminate intricate mechanisms of alveolar epithelial injury and recovery after blast trauma, highlighting autophagic and oxidative stress responses mediated by Thbs1-associated PI3K-Akt and p53 pathways as high-value therapeutic targets, and strategic modulation of these pathways in future studies may mitigate lung damage by reducing oxidative stress while engaging endogenous tissue repair processes like autophagy.
Collapse
Affiliation(s)
- Lin Zhang
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China; Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan 250001, China.
| | - Yongheng Wang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Linqiang Tian
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Long Li
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Zhen Chen
- School of Public Health, Weifang Medical University, Weifang 261000, China
| | - Chunjie Ding
- Anyang Tumor Hospital, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, Henan, China
| | - Jiaqi Tian
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan 250001, China
| | - Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan 250001, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang 453000, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China.
| |
Collapse
|
20
|
Bao R, Mao Y, Zhang Y, Chai J, Zhang Y, Luo C, Zhang K, Jiang G, He X. Fabrication of injectable alginate hydrogels with sustained release of 4-octyl itaconate for articular anti-inflammatory. Biomed Mater Eng 2024; 35:475-485. [PMID: 39150826 DOI: 10.3233/bme-240103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic and degenerative joint disease that remains a great challenge in treatment due to the lack of effective therapies. 4-octyl itaconate (4-OI) is a novel and potent modulator of inflammation for the treatment of inflammatory disease. However, the clinical usage of 4-OI is limited due to its poor solubility and low bioavailability. As a promising drug delivery strategy, injectable hydrogels offers an effective approach to address these limitations of 4-OI. OBJECTIVE The aim of the study was to verify that the composite 4-OI/SA hydrogels could achieve a controlled release of 4-OI and reduce damage to articular cartilage in the group of osteoarthritic rats treated with the system. METHODS In this study, an injectable composite hydrogel containing sodium alginate (SA) and 4-octyl itaconate (4-OI) has been developed for continuous intra-articular administration in the treatment of OA. RESULTS After intra-articular injection in arthritic rats, the as-prepared 4-OI/SA hydrogel containing of 62.5 μM 4-OI effectively significantly reduced the expression of TNF-α, IL-1β, IL-6 and MMP3 in the ankle fluid. Most importantly, the as-prepared 4-OI/SA hydrogel system restored the morphological parameters of the ankle joints close to normal. CONCLUSION 4-OI/SA hydrogel shows a good anti-inflammatory activity and reverse cartilage disruption, which provide a new strategy for the clinical treatment of OA.
Collapse
Affiliation(s)
- Ronghua Bao
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| | - Yifan Mao
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| | - Yuliang Zhang
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| | - Junlei Chai
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| | - Yuanbin Zhang
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| | - Cheng Luo
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| | - Kailong Zhang
- Department of Medical Research, Zhejiang Zhongwei Medical Research Co., Ltd, Hangzhou, China
| | - Guohua Jiang
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiaodan He
- Department of Orthopedics, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, China
| |
Collapse
|
21
|
You M, Jiang Q, Huang H, Ma F, Zhou X. 4-Octyl itaconate inhibits inflammation to attenuate psoriasis as an agonist of oxeiptosis. Int Immunopharmacol 2023; 124:110915. [PMID: 37741130 DOI: 10.1016/j.intimp.2023.110915] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023]
Abstract
Psoriasis is a highly prevalent chronic disease associated with a substantial social and economic burden. Oxeiptosis is a programmed cell death that occurs when cells are in a state of high oxidative stress, which has a potent anti-inflammatory effect. However, there is still no research on oxeiptosis in psoriasis, and the agonists or antagonists of oxeiptosis remain an unclear field. Here, we found that oxeiptosis of keratinocytes was inhibited in psoriasis lesions. KEAP1, as the upstream molecular component of oxeiptosis, is highly expressed in psoriasis lesions. Knockdown of KEAP1 in HaCaT cells caused oxeiptosis in the condition of M5 cocktail stimulation. Next, we found that the cell-permeable derivative of itaconate, 4-octylitaconate (OI) promoted oxeiptosis of keratinocytes by inhibiting KEAP1 and then activating PGAM5 which are two upstream molecular components of oxeiptosis. At the same time, OI can reduce the expression of inflammatory cytokines induced by M5 cocktail stimulation in vitro. Similarly, we found that OI can alleviate IMQ-induced psoriatic lesions in mice and downregulate the levels of inflammatory cytokines in psoriatic lesions. In summary, our findings suggest that oxeiptosis of keratinocytes was inhibited in psoriasis and OI can significantly inhibit inflammation and alleviate psoriasis as an agonist of oxeiptosis, indicating that oxeiptosis may be involved in regulating the progression of psoriasis, which may provide new therapeutic targets for psoriasis treatment.
Collapse
Affiliation(s)
- Mengshu You
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qian Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huining Huang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fangyu Ma
- Department of Health Management Center, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Xingchen Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
22
|
Sheng W, Wang Q, Qin H, Cao S, Wei Y, Weng J, Yu F, Zeng H. Osteoarthritis: Role of Peroxisome Proliferator-Activated Receptors. Int J Mol Sci 2023; 24:13137. [PMID: 37685944 PMCID: PMC10487662 DOI: 10.3390/ijms241713137] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023] Open
Abstract
Osteoarthritis (OA) represents the foremost degenerative joint disease observed in a clinical context. The escalating issue of population aging significantly exacerbates the prevalence of OA, thereby imposing an immense annual economic burden on societies worldwide. The current therapeutic landscape falls short in offering reliable pharmaceutical interventions and efficient treatment methodologies to tackle this growing problem. However, the scientific community continues to dedicate significant efforts towards advancing OA treatment research. Contemporary studies have discovered that the progression of OA may be slowed through the strategic influence on peroxisome proliferator-activated receptors (PPARs). PPARs are ligand-activated receptors within the nuclear hormone receptor family. The three distinctive subtypes-PPARα, PPARβ/δ, and PPARγ-find expression across a broad range of cellular terminals, thus managing a multitude of intracellular metabolic operations. The activation of PPARγ and PPARα has been shown to efficaciously modulate the NF-κB signaling pathway, AP-1, and other oxidative stress-responsive signaling conduits, leading to the inhibition of inflammatory responses. Furthermore, the activation of PPARγ and PPARα may confer protection to chondrocytes by exerting control over its autophagic behavior. In summation, both PPARγ and PPARα have emerged as promising potential targets for the development of effective OA treatments.
Collapse
Affiliation(s)
- Weibei Sheng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Qichang Wang
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Haotian Qin
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Siyang Cao
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yihao Wei
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jian Weng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Fei Yu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
23
|
Xu L, Cai J, Li C, Yang M, Duan T, Zhao Q, Xi Y, Sun L, He L, Tang C, Sun L. 4-Octyl itaconate attenuates LPS-induced acute kidney injury by activating Nrf2 and inhibiting STAT3 signaling. Mol Med 2023; 29:58. [PMID: 37095432 PMCID: PMC10127401 DOI: 10.1186/s10020-023-00631-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/08/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Septic acute kidney injury (S-AKI) is the leading form of acute kidney failure among hospitalized patients, and the inflammatory response is involved in this process. 4-octyl itaconate (4-OI) is a multi-target itaconate derivative with potent anti-inflammatory action. However, it remains elusive whether and how 4-OI contributes to the regulation of S-AKI. METHODS We employed a lipopolysaccharide (LPS)-induced AKI murine model and explored the potential renoprotective effect of 4-OI in vivo. In vitro experiments, BUMPT cells, a murine renal tubular cell line, were conducted to examine the effects of 4-OI on inflammation, oxidative stress, and mitophagy. Moreover, STAT3 plasmid was transfected in BUMPT cells to investigate the role of STAT3 signaling in the 4-OI-administrated state. RESULTS We demonstrate that 4-OI protects against S-AKI through suppressing inflammation and oxidative stress and enhancing mitophagy. 4-OI significantly reduced the levels of Scr, BUN, Ngal as well as the tubular injury in LPS-induced AKI mice. 4-OI restrained inflammation by reducing macrophage infiltration and suppressing the expression of IL-1β and NLRP3 in the septic kidney. 4-OI also reduced ROS levels, as well as cleaved caspase-3 and boosted antioxidants such as HO-1, and NQO1 in mice. In addition, the 4-OI treatment significantly promoted mitophagy. Mechanistically, 4-OI activated Nrf2 signaling and suppressed phosphorylated STAT3 in vivo and vitro. Molecular docking revealed the binding affinity of 4-OI towards STAT3. ML385, a specific Nrf2 inhibitor, partially repressed the anti-inflammatory and anti-oxidative effects of 4-OI and partially restricted the mitophagy induced by 4-OI in vivo and in vitro. Transfected with STAT3 plasmid partially suppressed mitophagy and the anti-inflammatory effect provoked by 4-OI in vitro. CONCLUSION These data suggest that 4-OI ameliorates LPS-induced AKI by suppressing inflammation and oxidative stress and enhancing mitophagy through the overactivation of the Nrf2 signaling pathway, and inactivation of STAT3. Our study identifies 4-OI as a promising pharmacologic for S-AKI.
Collapse
Affiliation(s)
- Lujun Xu
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Juan Cai
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Chenrui Li
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Ming Yang
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Tongyue Duan
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Qing Zhao
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Yiyun Xi
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Liya Sun
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Liyu He
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Chengyuan Tang
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China
| | - Lin Sun
- Department of Nephrology, Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, No.139 Renmin Middle Road, Changsha, Hunan, 410011, China.
| |
Collapse
|
24
|
Kim YJ, Park EJ, Lee SH, Silwal P, Kim JK, Yang JS, Whang J, Jang J, Kim JM, Jo EK. Dimethyl itaconate is effective in host-directed antimicrobial responses against mycobacterial infections through multifaceted innate immune pathways. Cell Biosci 2023; 13:49. [PMID: 36882813 PMCID: PMC9993662 DOI: 10.1186/s13578-023-00992-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Itaconate, a crucial immunometabolite, plays a critical role in linking immune and metabolic functions to influence host defense and inflammation. Due to its polar structure, the esterified cell-permeable derivatives of itaconate are being developed to provide therapeutic opportunities in infectious and inflammatory diseases. Yet, it remains largely uncharacterized whether itaconate derivatives have potentials in promoting host-directed therapeutics (HDT) against mycobacterial infections. Here, we report dimethyl itaconate (DMI) as the promising candidate for HDT against both Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria by orchestrating multiple innate immune programs. RESULTS DMI per se has low bactericidal activity against Mtb, M. bovis Bacillus Calmette-Guérin (BCG), and M. avium (Mav). However, DMI robustly activated intracellular elimination of multiple mycobacterial strains (Mtb, BCG, Mav, and even to multidrug-resistant Mtb) in macrophages and in vivo. DMI significantly suppressed the production of interleukin-6 and -10, whereas it enhanced autophagy and phagosomal maturation, during Mtb infection. DMI-mediated autophagy partly contributed to antimicrobial host defenses in macrophages. Moreover, DMI significantly downregulated the activation of signal transducer and activator of transcription 3 signaling during infection with Mtb, BCG, and Mav. CONCLUSION Together, DMI has potent anti-mycobacterial activities in macrophages and in vivo through promoting multifaceted ways for innate host defenses. DMI may bring light to new candidate for HDT against Mtb and nontuberculous mycobacteria, both of which infections are often intractable with antibiotic resistance.
Collapse
Affiliation(s)
- Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Chungbuk, South Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, South Korea
| | - Jeong Seong Yang
- Department of Research and Development, Korea Mycobacterium Resource Center (KMRC), The Korean Institute of Tuberculosis, Osong, 28158, South Korea
| | - Jake Whang
- Department of Research and Development, Korea Mycobacterium Resource Center (KMRC), The Korean Institute of Tuberculosis, Osong, 28158, South Korea
| | - Jichan Jang
- Division of Life Science, Department of Bio & Medical Big Data (BK21 Four Program), Research Institute of Life Science, Gyeongsang National University, Jinju, 52828, South Korea
| | - Jin-Man Kim
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.,Department of Pathology, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, South Korea. .,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, South Korea. .,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, South Korea.
| |
Collapse
|
25
|
Yu Z, Zhang Z, Zhang X, Bao J, Li H, Yu J, Shi N, Nan F, Cao L, Li C, Wang W. 4-Octyl itaconate treatment inhibits mitochondrial dysfunction and ER stress via stimulating SIRT1 expression in vitro and attenuates osteoarthritis process in murine DMM model in vivo. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
|
26
|
Xiao H, Dong Y, Wan D, Wan J, Huang J, Tang L, Zhou J, Yang T, Liu Y, Wang S. Injectable hydrogel loaded with 4-octyl itaconate enhances cartilage regeneration by regulating macrophage polarization. Biomater Sci 2023; 11:2445-2460. [PMID: 36757828 DOI: 10.1039/d2bm01894b] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Macrophages play a distinctive role in the early stage of inflammation after cartilage defects. Previous studies have shown that macrophages can express different phenotypes, among which M2 polarization is important to maintain the balance of the inflammatory microenvironment and promote cartilage regeneration. In this study, 4-octyl itaconic acid (4-OI), a derivative of the endogenous metabolite itaconic acid, was used to regulate the polarization behavior of macrophages and enhance cartilage repair. Oxidized sodium alginate (OSA) and gelatin (GEL) were selected as materials to form injectable hydrogels with the function of sustained release of 4-OI. In vivo and in vitro experiments have verified that the OSA/GEL hydrogel system loaded with 4-OI could promote M2 macrophage polarization and inhibit the inflammatory reaction. A rat knee joint cartilage defect model further confirmed its role in promoting cartilage regeneration in the later stage. In this study, the OSA/GEL hydrogel was successfully fabricated as a vehicle for delivering 4-OI, which could evidently alleviate the inflammatory reaction and thus accelerate tissue regeneration. The results of this study provide a new method for promoting subsequent tissue regeneration by regulating the early immune response.
Collapse
Affiliation(s)
- Hui Xiao
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Yunsheng Dong
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Dongdong Wan
- Department of Orthopedic, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| | - Jinpeng Wan
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Jiaxing Huang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Lizong Tang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Jie Zhou
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Tingting Yang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Yufei Liu
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
27
|
Mulberroside A alleviates osteoarthritis via restoring impaired autophagy and suppressing MAPK/NF-κB/PI3K-AKT-mTOR signaling pathways. iScience 2023; 26:105936. [PMID: 36698724 PMCID: PMC9868682 DOI: 10.1016/j.isci.2023.105936] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023] Open
Abstract
Osteoarthritis (OA) is a trauma-/age-related degenerative disease characterized by chronic inflammation as one of its pathogenic mechanisms. Mulberroside A (MA), a natural bioactive withanolide, demonstrates anti-inflammatory properties in various diseases; however, little is known about the effect of MA on OA. We aim to examine the role of MA on OA and to identify the potential mechanisms through which it protects articular cartilage. In vitro, MA improved inflammatory response, anabolism, and catabolism in IL-1β-induced OA chondrocytes. The chondroprotective effects of MA were attributed to suppressing the MAPK, NF-κB, and PI3K-AKT-mTOR signaling pathways, as well as promoting the autophagy process. In vivo, intra-articular injection of MA reduced the cartilage destruction and reversed the change of anabolic and catabolic-related proteins in destabilized medial meniscus (DMM)-induced OA models. Thus, the study indicates that MA exhibits a chondroprotective effect and might be a promising agent for OA treatment.
Collapse
|
28
|
Ma T, Wang X, Qu W, Yang L, Jing C, Zhu B, Zhang Y, Xie W. Osthole Suppresses Knee Osteoarthritis Development by Enhancing Autophagy Activated via the AMPK/ULK1 Pathway. Molecules 2022; 27:molecules27238624. [PMID: 36500713 PMCID: PMC9738845 DOI: 10.3390/molecules27238624] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Knee osteoarthritis (KOA) is an increasingly prevalent heterogeneous disease characterized by cartilage erosion and inflammation. As the main chemical constituent of Angelicae Pubescentis Radix (APR), an anti-inflammatory herbal medicine, the potential biological effects and underlying mechanism of osthole on chondrocytes and KOA progression remain elusive. In this study, the potential effect and mechanism of osthole on KOA were investigated in vitro and in vivo. We found that osthole inhibited IL-1β-induced apoptosis and cartilage matrix degeneration by activating autophagy in rat chondrocytes. In addition, osthole could activate autophagy through phosphorylation of AMPK/ULK1, and AMPK serves as a positive upstream regulator of ULK1. Furthermore, KOA rats treated with osthole showed phosphorylation of the AMPK/ULK1 pathway and autophagy activation, as well as cartilage protection. Collectively, the AMPK/ULK1 signaling pathway can be activated by osthole to enhance autophagy, thereby suppressing KOA development. Osthole may be a novel and effective therapeutic agent for the clinical treatment of KOA.
Collapse
Affiliation(s)
- Teng Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiangpeng Wang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Wenjing Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lingsen Yang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Cheng Jing
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Bingrui Zhu
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Yongkui Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
- Correspondence: (Y.Z.); (W.X.)
| | - Wenpeng Xie
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
- Correspondence: (Y.Z.); (W.X.)
| |
Collapse
|
29
|
Chen Y, Pan X, Zhao J, Li C, Lin Y, Wang Y, Liu X, Tian M. Icariin alleviates osteoarthritis through PI3K/Akt/mTOR/ULK1 signaling pathway. Eur J Med Res 2022; 27:204. [PMID: 36253872 PMCID: PMC9575285 DOI: 10.1186/s40001-022-00820-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES This study aims to investigate the effects of Icariin (ICA) on interleukin-1β (IL-1β)-induced osteoarthritis (OA) and its potential mechanism of action. METHODS SW1353 chondrocytes were pretreated with ICA for 2 h, followed by stimulation with IL-1β to mimic OA. Expression levels of matrix metalloproteinases (MMP-3) and collagen II were determined using real-time PCR and Western blot assays. Autophagy activation (by ICA) or inhibition (by shRNA) was determined based on the expression levels of ULK1, Beclin-1, LC3-II/I, and p62, using Western blot analysis. The phosphorylation levels of PI3K, Akt, mTOR, and ULK1 were also detected using Western blot analysis. RESULTS IL-1β increased MMP-3 overproduction, induced collagen II degradation, and reduced the level of autophagy-associated proteins, including ULK1, Beclin-1, and LC3-II/I. In contrast, ICA pretreatment attenuated IL-1β-induced MMP-3 overproduction, increased collagen II expression, and induced expression of autophagy-related proteins. ICA also decreased PI3K, Akt, and mTOR phosphorylation, increased the production of ULK1, and induced autophagy. Short hairpin RNA-mediated knockdown of ULK1 led to activation of the PI3K/Akt/mTOR pathway, which reversed the protective effects of ICA. CONCLUSIONS Our findings indicate that ICA can induce autophagy by regulating the PI3K/AKT/mTOR/ULK1 signaling pathway. This study suggests that ICA may be effective for treating OA.
Collapse
Affiliation(s)
- Yan Chen
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Xiaoli Pan
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
| | - Jing Zhao
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Chunyan Li
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Yupei Lin
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Yu Wang
- Department of Rheumatology and Immunology Department, Zunyi Medical University, Zunyi, 563006, China
| | - Xu Liu
- Department of Rheumatology and Immunology Department, Peking University People's Hospital, Beijing, 100044, China
| | - Mei Tian
- Department of Rheumatology and Immunology Department, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China.
| |
Collapse
|
30
|
tRNA-Derived Fragment tRF-5009A Regulates Autophagy and Degeneration of Cartilage in Osteoarthritis via Targeting mTOR. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5781660. [PMID: 36035226 PMCID: PMC9410839 DOI: 10.1155/2022/5781660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022]
Abstract
tRNA-derived fragments (tRFs) have been reported to have critical regulatory roles in osteoarthritis (OA). Recent studies have suggested that autophagy promotes the homeostasis of the extracellular matrix of chondrocytes in OA. However, the role of tRFs in posttranscriptional gene regulation during autophagy in OA is unknown. Therefore, we explored the role of tRF-5009A in the posttranscriptional gene regulation of autophagy and cartilage degeneration in OA. Using RNA sequencing, we identified tRF-5009A, the tRNAValCAC-derived fragment, in OA tissues and explored its expression by quantitative reverse transcription PCR and fluorescence in situ hybridization. We further investigated the relationship between the expression of tRF-5009A and clinical factors in OA. Chondrocytes were transfected with a tRF-5009A inhibitor or mimic to determine their functions, including in relation to autophagy and the cartilage phenotype. A rescue experiment and dual-luciferase reporter assay were conducted to determine whether the 3′-untranslated region (UTR) of mTOR contains a tRF-5009A-binding site. tRF-5009A was downregulated in the cartilage of OA knees, especially in damaged areas. mTOR was highly expressed in damaged cartilage and negatively correlated with the expression of tRF-5009A; transfection with a tRF-5009A inhibitor promoted the expression of mTOR and suppressed autophagy, whereas transfection with a tRF-5009A mimic had the opposite effect. A dual-luciferase reporter assay showed that tRF-5009A silenced the expression of mTOR by binding to its 3′-UTR. Thus, tRF-5009A regulates autophagy and cartilage degeneration in OA by targeting mTOR. In summary, these findings provide an additional tool for the clinical diagnosis and novel targeted therapy of OA.
Collapse
|