1
|
Lu J, Cai J, Zhou Z, Ma J, Han T, Lu N, Zhu L. Gel@CAT-L hydrogel mediates mitochondrial unfolded protein response to regulate reactive oxygen species and mitochondrial homeostasis in osteoarthritis. Biomaterials 2025; 321:123283. [PMID: 40222260 DOI: 10.1016/j.biomaterials.2025.123283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE This study investigates the role of Gelatin-Catalase (Gel@CAT)-L hydrogel in mediating reactive oxygen species (ROS) production and maintaining mitochondrial homeostasis through SIRT3-mediated unfolded protein response (UPRmt), while exploring its involvement in the molecular mechanism of osteoarthritis (OA). METHODS Self-assembled Gel@CAT-L hydrogels were fabricated and characterized using transmission electron microscopy, mechanical testing, external release property evaluation, and oxygen production measurement. Biocompatibility was assessed via live/dead cell staining and CCK8 assays. An OA mouse model was established using destabilization of the medial meniscus (DMM) surgery. X-ray and micro-CT imaging were employed to evaluate the structural integrity of the mouse knee joints, while histological staining was used to assess cartilage degeneration. Immunohistochemistry was performed to analyze the expression of proteins including Col2a1, Aggrecan, MMP13, ADAMTS5, SIRT3, PINK1, and Parkin. Multi-omics analyses-encompassing high-throughput sequencing, proteomics, and metabolomics-were conducted to identify key genes and metabolic pathways targeted by Gel@CAT-L hydrogel intervention in OA. Immunofluorescence techniques were utilized to measure ROS levels, mitochondrial membrane potential, and the expression of SIRT3, PINK1, Parkin, LYSO, LC3B, Col2a1, and MMP13 in primary mouse chondrocytes and mouse knee joints. Flow cytometry was applied to quantify ROS-positive cells. RT-qPCR analysis was conducted to determine mRNA levels of Aggrecan, Col2a1, ADAMTS5, MMP13, SIRT3, mtDNA, HSP60, LONP1, CLPP, and Atf5 in primary mouse chondrocytes, mouse knee joints, and human knee joints. Western blotting was performed to measure protein expression levels of SIRT3, HSP60, LONP1, CLPP, and Atf5 in both primary mouse chondrocytes and mouse knee joints. Additionally, 20 samples each from the control (CON) and OA groups were collected for analysis. Hematoxylin and eosin staining was used to evaluate cartilage degeneration in human knee joints. The Mankin histological scoring system quantified the degree of cartilage degradation, while immunofluorescence analyzed SIRT3 protein expression in human knee joints. RESULTS In vitro experiments demonstrated that self-assembled Gel@CAT-L hydrogels exhibited excellent biodegradability and oxygen-releasing capabilities, providing a stable three-dimensional environment conducive to cell viability and proliferation while reducing ROS levels. Multi-omics analysis identified SIRT3 as a key regulatory gene in mitigating OA and revealed its central role in the UPRmt pathway. Furthermore, Gel@CAT-L was confirmed to regulate mitochondrial homeostasis. Both in vitro experiments and in vivo mouse model studies confirmed that Gel@CAT-L significantly reduced ROS levels and regulated mitochondrial autophagy by activating the SIRT3-mediated UPRmt pathway, thereby improving the pathological state of OA. Clinical trials indicated downregulation of SIRT3 and UPRmt-related proteins in OA patients. CONCLUSION Gel@CAT-L hydrogel activates SIRT3-mediated UPRmt to regulate ROS and mitochondrial homeostasis, providing potential therapeutic benefits for OA.
Collapse
Affiliation(s)
- Jiajia Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Jiao Cai
- Department of Medical Administration, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Zhibin Zhou
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China
| | - Jun Ma
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China; Department of Orthopaedic Trauma, Naval Medical Center of PLA, Naval Medical University, Shanghai, 200001, China
| | - Tianyu Han
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China.
| | - Nan Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| | - Lei Zhu
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China.
| |
Collapse
|
2
|
Liu J, Liu J, Liu S, Xiao P, Du C, Zhan J, Chen Z, Chen L, Li K, Huang W, Lei Y. Cascade targeting selenium nanoparticles-loaded hydrogel microspheres for multifaceted antioxidant defense in osteoarthritis. Biomaterials 2025; 318:123195. [PMID: 39965424 DOI: 10.1016/j.biomaterials.2025.123195] [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: 11/28/2024] [Revised: 02/01/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Selenium (Se) deficiency is a critical factor contributing to the imbalance of redox homeostasis in chondrocytes and the progression of osteoarthritis (OA). However, traditional selenium supplements face challenges such as a narrow therapeutic window and lack of targeting. To address this, we designed hyaluronic acid (HA)-modified selenium nanoparticles (HA-SeNPs) and developed a cascade-targeted delivery system (HA-SeNPs@AHAMA-HMs) based on a nano-micron combined strategy. The system involves loading HA-SeNPs into aldehyde-functionalized hydrogel microspheres prepared via microfluidic technology. Through Schiff base reactions between the aldehyde groups of the microspheres and amino groups of the cartilage, the system selectively adheres to the surface of damaged cartilage, achieving micron-scale targeting while continuously releasing HA-SeNPs. Then, HA-SeNPs achieve nanoscale targeting by binding to CD44, which is highly expressed on OA chondrocyte membranes, via their HA surface. Once taken up by the cells, HA-SeNPs exert their effects by directly scavenging ROS and promoting selenoprotein synthesis through the generation of selenite, forming a multifaceted antioxidant defense system. This effectively alleviates oxidative stress and optimizes mitochondrial function. In vivo and in vitro results demonstrated that this system significantly improved the oxidative phosphorylation pathway associated with mitochondrial function, which markedly reduced joint space narrowing and cartilage matrix degradation, and delayed the progression of OA. In summary, this study suggests that the cascade-targeting hydrogel microspheres designed and constructed based on a nano-micron combined strategy represent a promising prospective approach for precise Se supplementation and OA treatment.
Collapse
Affiliation(s)
- Jiacheng Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Junyan Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Chengcheng Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Jingdi Zhan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Lu Chen
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ke Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Yiting Lei
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing, 400016, China; Department of Biomedical Engineering, The Chinese University of Hong Kong, NT, Hong Kong SAR, 999077, China.
| |
Collapse
|
3
|
Xiong J, Guo Q, Luo X. Cellular senescence in age-related musculoskeletal diseases. Front Med 2025:10.1007/s11684-025-1125-7. [PMID: 40314896 DOI: 10.1007/s11684-025-1125-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/16/2024] [Indexed: 05/03/2025]
Abstract
Aging is typically associated with decreased musculoskeletal function, leading to reduced mobility and increased frailty. As a hallmark of aging, cellular senescence plays a crucial role in various age-related musculoskeletal diseases, including osteoporosis, osteoarthritis, intervertebral disc degeneration, and sarcopenia. The detrimental effects of senescence are primarily due to impaired regenerative capacity of stem cells and the pro-inflammatory environment created by accumulated senescent cells. The secreted senescence-associated secretory phenotype (SASP) can induce senescence in neighboring cells, further amplifying senescent signals. Although the removal of senescent cells and the suppression of SASP factors have shown promise in alleviating disease progression and restoring musculoskeletal health in mouse models, clinical trials have yet to demonstrate significant efficacy. This review summarizes the mechanisms of cellular senescence in age-related musculoskeletal diseases and discusses potential therapeutic strategies targeting cellular senescence.
Collapse
Affiliation(s)
- Jinming Xiong
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Qiaoyue Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| |
Collapse
|
4
|
Zhou S, Liao F, Wen H. Isoquercetin alleviates osteoarthritis via regulating the NOX4/Nrf2 redox imbalance in senescent chondrocytes. Int J Biol Macromol 2025; 306:141562. [PMID: 40020820 DOI: 10.1016/j.ijbiomac.2025.141562] [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: 11/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The redox imbalance induced by excessive reactive oxygen species (ROS) contributes to senescent phenotypes of chondrocytes in osteoarthritis (OA). However, there is limited evidence regarding the involvement of NADPH oxidase-4 (NOX4)/NFE2-related factor 2 (Nrf2) redox imbalance in OA. Isoquercetin (IQ), a quercetin derivative, exhibits promising antioxidative and anti-aging properties. Here, we found that IQ promoted redox homeostasis by inhibiting NOX4 and activating Nrf2-mediated antioxidant responses, thereby ameliorating OA. Specifically, IQ significantly suppressed the expression of senescence-associated secretory phenotypes (SASPs) in senescent chondrocytes. RNA sequencing analysis revealed that cellular senescence and oxidative stress were involved in the mechanism of IQ's effect on senescent chondrocytes. IQ effectively reversed redox imbalance, as evidenced by reduced levels of ROS and endogenous oxidants, and increased mitochondrial membrane potential and, elevated levels of endogenous antioxidants. Mechanistically, the elevated expression of NOX4 observed in patients with severe OA confirms its role in OA pathogenesis. Molecular docking and NOX4 knockdown experiments suggested that IQ may interact with NOX4 and inhibit its expression. This study identifies a potential therapeutic target and provides a promising candidate for OA treatment.
Collapse
Affiliation(s)
- Siqi Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Haiyan Wen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
5
|
Chen X, Ma T, Chen Y, Sun Q, Wang H, Wang Y. USP14 promotes osteoarthritis progression by deubiquitinating FZD8 to activate the Wnt/β-catenin signaling pathway. Immunobiology 2025; 230:152905. [PMID: 40318610 DOI: 10.1016/j.imbio.2025.152905] [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: 01/22/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease and associated with multiple pathogenic factors, such as old age, heredity, obesity, mechanical damage and inflammatory gene mutation. In this study, we aimed to explore the functions of ubiquitin specific peptidase 14 (USP14) in OA development. METHODS The in vitro model of OA was constructed by stimulating chondrocytes with IL-1β. qRT-PCR and western blot assays were used for gene expression. MTT assay and EdU assay were manipulated to evaluate cell proliferation. Flow cytometry analysis was conducted for cell apoptosis. ELISA kits were utilized to determine the concentrations of inflammatory cytokines. Co-immunoprecipitation (Co-IP) assay and GST pull-down assay were manipulated to estimate the interaction between USP14 and Frizzled 8 (FZD8). Ubiquitination assay was used to evaluate the deubiquitination of FZD8. RESULTS USP14 was highly expression in OA cartilage tissues and IL-1β-triggered chondrocytes. USP14 silencing aggravated the proliferation and repressed the apoptosis, inflammation and extracellular matrix (ECM) degradation of IL-1β-treated chondrocytes. USP14 could interact with FZD8 and regulate FZD8 expression through FZD8 deubiquitination. Moreover, FZD8 overexpression alleviated the effects of UPS14 silencing on IL-1β-treated chondrocyte proliferation, apoptosis, inflammation and ECM degradation. Additionally, USP14 knockdown inhibited Wnt/β-catenin signal pathway via the deubiquitination of FZD8. CONCLUSION USP14 repressed IL-1β-treated chondrocyte proliferation and promoted apoptosis, inflammation and ECM degradation by regulating FZD8 expression and Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Xiaochao Chen
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Tiancheng Ma
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Yongfeng Chen
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Qiang Sun
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Huayi Wang
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China
| | - Yuanrui Wang
- Department of Orthopaedics, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, China..
| |
Collapse
|
6
|
Sun H, Xu C, Xiong Z, Liu M, Ning X, Zhuang Y. Therapeutic prospects and potential mechanisms of Prdx6: as a novel target in musculoskeletal disorders. Front Physiol 2025; 16:1524100. [PMID: 40313876 PMCID: PMC12043587 DOI: 10.3389/fphys.2025.1524100] [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: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
With the global population aging, musculoskeletal disorders (MSDs) have posed significant physical and psychological health challenges for patients as well as a substantial economic burden on society. The advancements in conservative and surgical interventions for MSDs have been remarkable in recent years; however, the current treatment modalities still fall short of meeting the optimal requirements of patients. Recently, peroxiredoxin 6 (Prdx6) has gained considerable attention from researchers due to its remarkable antioxidative, anti-inflammatory, and anti-apoptotic properties. It has been found that Prdx6 is involved in multiple system diseases, including MSDs; however, the exact role of Prdx6 in MSDs is still lacking. This study aimed to summarize the structure, regulatory mechanism, and potential function of Prdx6. These findings may demonstrate Prdx6 as a novel target for inhibiting the advancement of MSDs.
Collapse
Affiliation(s)
- Hong Sun
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Xu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhilin Xiong
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Miao Liu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xu Ning
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong Zhuang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
7
|
Zhang X, Fang Z, Heng BC, Hu X, Ge Z. Mitigating Oxidative Stress Enhances Cartilage Regeneration by Ameliorating Apoptosis of Cartilage Progenitor Cells in Adult Mice. Adv Biol (Weinh) 2025:e2400739. [PMID: 40110588 DOI: 10.1002/adbi.202400739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/17/2025] [Indexed: 03/22/2025]
Abstract
Cartilage regeneration in juvenile mice was better than in adult mice. This study evaluated the roles of cytokines and reactive oxygen species (ROS) after cartilage injury in both juvenile and adult mice and attempted to correlate these with cartilage progenitor cells and age-related differences in cartilage regeneration. Full-thickness cartilage defects were created in the femoral trochlea of knee joints in both 4-week-old (juvenile) and 8-week-old (adult) mice. Adult mice showed higher ROS peaks than juveniles at day 7 post-injury. Protein expression levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) significantly decreased after surgery, while interleukin-1β (IL-1β) levels displayed no significant change. There were more cartilage progenitor cells together with more cell proliferation in juvenile versus adult mice, while there was much less apoptosis of cartilage progenitor cells in juvenile mice compared to adult mice. ROS inhibition enhanced cartilage regeneration in adult mice by promoting progenitor cell proliferation and reducing apoptosis, mimicking the regenerative pattern seen in juveniles. This study demonstrated that inhibiting ROS in adult mice promoted cartilage regeneration, possibly by enhancing proliferation and decreasing apoptosis of cartilage progenitor cells.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, P. R. China
| | - Zhihao Fang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, P. R. China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, 100084, P. R. China
- Tsinghua-Peking Centre for Life Sciences, Beijing, 100084, P. R. China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, 100084, P. R. China
| | - Zigang Ge
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, P. R. China
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, P. R. China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| |
Collapse
|
8
|
Cao X, Yang X, Zhang P, Xu J, Zhao J, Yang E. Targeting Txnip-mediated metabolic reprogramming has therapeutic potential for osteoarthritis. Cell Death Discov 2025; 11:110. [PMID: 40113744 PMCID: PMC11926230 DOI: 10.1038/s41420-025-02394-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/13/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Osteoarthritis (OA) inflammatory microenvironment triggered glucose metabolism and mitochondrial dysfunction in chondrocytes, leading to a shift of metabolic tendency between oxidative phosphorylation and anaerobic glycolysis. Thioredoxin-interacting protein (Txnip) increased production of reactive oxygen species (ROS), which exacerbates oxidative stress, inflammation and further accelerates cartilage degeneration and extracellular matrix (ECM) degradation. Txnip expression is also positively correlated with several critical pathological glucose and lipid metabolism processes beyond inflammation and endoplasmic reticulum stress (ERS). While the role of Txnip-mediated chondrocyte metabolic reprogramming in OA has not been explored. This study focuses on the unexplored role of Txnip-mediated chondrocyte metabolic reprogramming in chondrogenesis and ECM deposition. The study reveals that upregulated glycolysis after Txnip knockdown significantly contributes to mouse chondrogenesis and ECM deposition. Moreover, verapamil, a clinically used drug that targets Txnip, shows potential for treating mouse OA. These findings suggest that targeting Txnip-mediated metabolic reprogramming could offer a novel therapeutic strategy for OA treatment.
Collapse
Affiliation(s)
- Xiankun Cao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Xiao Yang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Pu Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China
| | - Jianguang Xu
- Department of Orthopedics Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Erzhu Yang
- Department of Orthopedics Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| |
Collapse
|
9
|
Li N, Zhang Z, Shen L, Song G, Tian J, Liu Q, Ni J. Selenium metabolism and selenoproteins function in brain and encephalopathy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:628-656. [PMID: 39546178 DOI: 10.1007/s11427-023-2621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/09/2024] [Indexed: 11/17/2024]
Abstract
Selenium (Se) is an essential trace element of the utmost importance to human health. Its deficiency induces various disorders. Se species can be absorbed by organisms and metabolized to hydrogen selenide for the biosynthesis of selenoproteins, selenonucleic acids, or selenosugars. Se in mammals mainly acts as selenoproteins to exert their biological functions. The brain ranks highest in the specific hierarchy of organs to maintain the level of Se and the expression of selenoproteins under the circumstances of Se deficiency. Dyshomeostasis of Se and dysregulation of selenoproteins result in encephalopathy such as Alzheimer's disease, Parkinson's disease, depression, amyotrophic lateral sclerosis, and multiple sclerosis. This review provides a summary and discussion of Se metabolism, selenoprotein function, and their roles in modulating brain diseases based on the most currently published literature. It focuses on how Se is utilized and transported to the brain, how selenoproteins are biosynthesized and function physiologically in the brain, and how selenoproteins are involved in neurodegenerative diseases. At the end of this review, the perspectives and problems are outlined regarding Se and selenoproteins in the regulation of encephalopathy.
Collapse
Affiliation(s)
- Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Guoli Song
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Jiazuan Ni
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| |
Collapse
|
10
|
Zhou J, Obianwuna UE, Zhang L, Liu Y, Zhang H, Qiu K, Wang J, Qi G, Wu S. Comparative effects of selenium-enriched lactobacilli and selenium-enriched yeast on performance, egg selenium enrichment, antioxidant capacity, and ileal microbiota in laying hens. J Anim Sci Biotechnol 2025; 16:27. [PMID: 39966907 PMCID: PMC11837603 DOI: 10.1186/s40104-025-01160-6] [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: 10/16/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Organic selenium (Se) has gained recognition in poultry nutrition as a feed additive to boost production and Se deposition in eggs and tissues, owing to its high bioavailability, efficient tissue accumulation and minimal toxicity. Selenium-enriched yeast (SeY) is a well-established source, while selenium-enriched lactobacilli (SeL), a newer alternative, offers the added benefits of probiotics. This study examined the effects of SeY and SeL on egg quality, antioxidant capacity, Se deposition, and gut health in laying hens. After a two-week pre-treatment with a Se-deficient diet (SeD), 450 Hy-Line Brown laying hens (30-week-old) were assigned into five dietary groups with six replicates of 15 hens each. The groups included a SeD, SeD supplemented with 1.5 mg Se/kg from SeY (SeY15), or 1.5, 3.0, and 6.0 mg Se/kg from SeL (SeL15, SeL30, SeL60). The feeding trial lasted for 12 weeks. RESULTS SeY15 and SeL15 improved the feed-to-egg ratio (P < 0.05) in the latter stages. Haugh units were significantly increased (P < 0.05) in the SeY15 and SeL30 groups, while darker yolk color (P < 0.05) was observed in the SeY15, SeL15, and SeL60 groups. All Se-supplemented diets increased Se content in whole eggs, albumen, and yolk (P < 0.05), while SeL groups showed a dose-dependent effect. Antioxidant enzyme activities increased, and MDA content decreased in the serum (P < 0.05), with SeY15 showing the highest GSH-Px levels (P < 0.05). SeL60 increased serum alkaline phosphatase and aspartate transaminase, and distorted the liver architecture (P < 0.05). Se-diets reduced concentrations of reactive oxygen species (ROS) in the ileum and liver (P < 0.05). SeL15 improved the ileal villus height-to-crypt depth ratio (P < 0.05). SeY15 and/or SeL15 up-regulated TXNRD1 and SEPHS1 mRNA while down-regulating SCLY expression in the liver. SeY15 altered ileal microbiota by increasing both beneficial and pathogenic bacteria, whereas SeL15 predominantly boosted beneficial bacteria. CONCLUSION SeL integrates the antioxidant properties of organic Se with the probiotic benefits on gut health, resulting in a performance-enhancing effect comparable to that of SeY. However, high SeL level (6.0 mg Se/kg) compromised productivity and metabolic functions while enhancing Se deposition.
Collapse
Affiliation(s)
- Jianmin Zhou
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Uchechukwu Edna Obianwuna
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Longfei Zhang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- College of Animal Science and Technology, Beijing Agricultural University, Beijing, 100096, China
| | - Yongli Liu
- Baiyian Biological Engineering Co., Ltd., Jiaozuo, Henan, 454000, China
| | - Haijun Zhang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Kai Qiu
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Jing Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Guanghai Qi
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
| | - Shugeng Wu
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, China Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China.
| |
Collapse
|
11
|
Batyrova G, Taskozhina G, Umarova G, Umarov Y, Morenko M, Iriskulov B, Kudabayeva K, Bazargaliyev Y. Unveiling the Role of Selenium in Child Development: Impacts on Growth, Neurodevelopment and Immunity. J Clin Med 2025; 14:1274. [PMID: 40004804 PMCID: PMC11856779 DOI: 10.3390/jcm14041274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Selenium (Se) is a vital trace element for children, playing a crucial role in numerous physiological processes, including antioxidant defense, immune regulation, thyroid function, and bone metabolism. Emerging evidence highlights its potential impact on child development and growth while also underscoring the complexity of its mechanisms and the global variations in Se intake. The aim of this review is to comprehensively elucidate the significance of Se in various biological processes within the human body, with a focus on its role in child development and growth; its biochemical effects on the nervous system, thyroid function, immune system, and bone tissue; and the implications of Se deficiency and toxicity. This review integrates findings from experimental models, epidemiological studies, and clinical trials to explore Se's role in neurodevelopment, growth regulation, and immune competence in children. Selenoproteins, which regulate oxidative stress and thyroid hormone and bone metabolism, are essential for normal growth and cognitive development in children. Se deficiency and toxicity has been linked to impaired immune function, growth retardation, and decreased immune function. The findings underscore Se's influence on various biological pathways that are critical for healthy child development and its broader importance for child health. Public health strategies aimed at optimizing selenium intake may play a pivotal role in improving pediatric health outcomes worldwide.
Collapse
Affiliation(s)
- Gulnara Batyrova
- Department of Clinical Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Gulaim Taskozhina
- Department of Clinical Laboratory Diagnostics, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Gulmira Umarova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan
| | - Yeskendir Umarov
- Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Marina Morenko
- Department of Children’s Diseases, Astana Medical University, Astana 010000, Kazakhstan;
| | - Bakhtiyar Iriskulov
- Department of Normal and Pathological Physiology, Tashkent Medical Academy, Tashkent 100109, Uzbekistan;
| | - Khatimya Kudabayeva
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (K.K.); (Y.B.)
| | - Yerlan Bazargaliyev
- Department of Internal Diseases 1, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (K.K.); (Y.B.)
| |
Collapse
|
12
|
Kang D, Lee J, Yook G, Jeong S, Shin J, Kim MS, Kim YJ, Jung H, Ahn J, Kim TW, Chang MJ, Chang CB, Kang SB, Yang WH, Lee YH, Cho JW, Yi EC, Kang C, Kim JH. Regulation of senescence-associated secretory phenotypes in osteoarthritis by cytosolic UDP-GlcNAc retention and O-GlcNAcylation. Nat Commun 2025; 16:1094. [PMID: 39904978 PMCID: PMC11794700 DOI: 10.1038/s41467-024-55085-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/27/2024] [Indexed: 02/06/2025] Open
Abstract
UDP-GlcNAc serves as a building block for glycosaminoglycan (GAG) chains in cartilage proteoglycans and simultaneously acts as a substrate for O-GlcNAcylation. Here, we show that transporters for UDP-GlcNAc to the endoplasmic reticulum (ER) and Golgi are significantly downregulated in osteoarthritic cartilage, leading to increased cytosolic UDP-GlcNAc and O-GlcNAcylation in chondrocytes. Mechanistically, upregulated O-GlcNAcylation governs the senescence-associated secretory phenotype (SASP) by stabilizing GATA4 via O-GlcNAcylation at S406, which compromises its degradation by p62-mediated selective autophagy. Elevated O-GlcNAcylation in the superficial layer of osteoarthritic cartilage coincides with increased GATA4 levels. The topical deletion of Gata4 in this cartilage layer ameliorates post-traumatic osteoarthritis (OA) in mice while inhibiting O-GlcNAc transferase mitigates OA by decreasing GATA4 levels. Excessive glucosamine-induced O-GlcNAcylation stabilizes GATA4 in chondrocytes and exacerbates post-traumatic OA in mice. Our findings elucidate the role of UDP-GlcNAc compartmentalization in regulating secretory pathways associated with chronic joint inflammation, providing a senostatic strategy for the treatment of OA.
Collapse
Affiliation(s)
- Donghyun Kang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea
| | - Jeeyeon Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea
| | - Geunho Yook
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea
| | - Sehan Jeong
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea
| | - Jungkwon Shin
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea
| | - Mi-Sung Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Yi-Jun Kim
- Department of Environmental Medicine, College of Medicine, Ewha Womans University, Seoul, 07985, South Korea
| | - Hyeryeon Jung
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
| | - Jinsung Ahn
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
| | - Tae Woo Kim
- Department of Orthopaedic Surgery, Seoul National University Boramae Hospital, Seoul, 07061, South Korea
| | - Moon Jong Chang
- Department of Orthopaedic Surgery, Seoul National University Boramae Hospital, Seoul, 07061, South Korea
| | - Chong Bum Chang
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Seung-Baik Kang
- Department of Orthopaedic Surgery, Seoul National University Boramae Hospital, Seoul, 07061, South Korea
| | - Won Ho Yang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
- Glycosylation Network Research Center, Yonsei University, Seoul, 03722, South Korea
| | - Yong-Ho Lee
- Glycosylation Network Research Center, Yonsei University, Seoul, 03722, South Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jin Won Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
- Glycosylation Network Research Center, Yonsei University, Seoul, 03722, South Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
- Glycosylation Network Research Center, Yonsei University, Seoul, 03722, South Korea
| | - Chanhee Kang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jin-Hong Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea.
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.
- Glycosylation Network Research Center, Yonsei University, Seoul, 03722, South Korea.
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang, 25354, South Korea.
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
13
|
Qi X, Tong L, Lian H, Chen Z, Yang L, Wu Y, Jin S, Guo D. Selenium nanoparticles modified with Ophiocordyceps gracilis polysaccharides: Enhancing stability, bioavailability, and anti-inflammatory efficacy. Food Res Int 2025; 201:115652. [PMID: 39849788 DOI: 10.1016/j.foodres.2024.115652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/25/2025]
Abstract
Here, a high molecular weight polysaccharide preparation from Ophiocordyceps gracilis was utilized as a stabilizer and dispersant to create nanocomposites based on selenium nanoparticles (GSP-1a-SeNPs). The NPs showed the highest stability at a selenium/polysaccharide mass ratio of 1:1, with no significant change after 28 days of storage at 4 °C. The NPs exhibited a symmetrical spheroid structure with an average diameter of 85.4 nm. Next, the anti-inflammatory properties and mechanisms of the GSP-1a-SeNPs were examined in LPS-induced RAW264.7 cells, which efficiently internalized the NPs. In the anti-inflammatory assays, GSP-1a-SeNPs significantly reduced the production of pro-inflammatory cytokines, including TNF-α and IL-6, and lowered ROS levels by activating the Nrf2-Keap1 pathway. This pathway regulates selenoprotein expression, thereby balancing the immune microenvironment of RAW264.7 cells and mitigating inflammation. These results suggest that GSP-1a-SeNPs could serve as potential therapeutic agents or adjuvants for treating LPS-induced inflammation.
Collapse
Affiliation(s)
- Xinya Qi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Lingling Tong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Hui Lian
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Zilei Chen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Linhui Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yang Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Suxing Jin
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Dongsheng Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
14
|
Wang Q, Feng K, Wan G, Liao W, Jin J, Wang P, Sun X, Wang W, Jiang Q. A ROS-responsive hydrogel encapsulated with matrix metalloproteinase-13 siRNA nanocarriers to attenuate osteoarthritis progression. J Nanobiotechnology 2025; 23:18. [PMID: 39815302 PMCID: PMC11737235 DOI: 10.1186/s12951-024-03046-7] [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: 07/04/2024] [Accepted: 11/29/2024] [Indexed: 01/30/2025] Open
Abstract
RNA interference (RNAi) and oxidative stress inhibition therapeutic strategies have been extensively utilized in the treatment of osteoarthritis (OA), the most prevalent degenerative joint disease. However, the synergistic effects of these approaches on attenuating OA progression remain largely unexplored. In this study, matrix metalloproteinase-13 siRNA (siMMP-13) was incorporated onto polyethylenimine (PEI)-polyethylene glycol (PEG) modified Fe3O4 nanoparticles, forming a nucleic acid nanocarrier termed si-Fe NPs. Subsequently, a poly(vinyl alcohol) (PVA) crosslinked phenylboronic acid (PBA)-modified hyaluronic acid (HA) hydrogel (HPP) was used to encapsulate the si-Fe NPs, resulting in a bifunctional hydrogel (si-Fe-HPP) with reactive oxygen species (ROS)-responsive and RNAi therapeutic properties. Studies in vitro demonstrated that si-Fe-HPP exhibited excellent biocompatibility, anti-inflammatory effects and prolonged stable retention time in knee joint. Intra-articular injection of si-Fe-HPP significantly attenuated cartilage degradation in mice with destabilization of the medial meniscus (DMM)-induced OA. The si-Fe-HPP treatment not only notably alleviated synovitis, osteophyte formation and subchondral bone sclerosis, but also markedly improved physical activity and reduced pain in DMM-induced OA mice. This study reveals that si-Fe-HPP, with its ROS-responsive and RNAi abilities, can significantly protect chondrocytes and attenuate OA progression, providing novel insights and directions for the development of therapeutic materials for OA treatment.
Collapse
Affiliation(s)
- Qiuyang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, 210008, People's Republic of China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People's Republic of China
| | - Kai Feng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Guangsheng Wan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Wei Liao
- Children's Hospital of Nanjing Medical University, Nanjing, 210008, People's Republic of China
| | - Jing Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, 210008, People's Republic of China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People's Republic of China.
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Weijun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, 210008, People's Republic of China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People's Republic of China.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China.
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, 210008, People's Republic of China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, People's Republic of China.
| |
Collapse
|
15
|
Li L, Zhu J, Chen Y, Li H, Han Y, Zhang L, Wang B. Interaction Between YTH Domain-Containing Family Protein 2 and SET Domain-Containing Lysine Methyltransferase 7 Suppresses Autophagy in Osteoarthritis Chondrocytes, Exacerbating Cartilage Damage. J Gene Med 2025; 27:e70005. [PMID: 39789715 DOI: 10.1002/jgm.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/31/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Osteoarthritis (OA) is characterized by progressive cartilage degeneration mediated by various molecular pathways, including inflammatory and autophagic processes. SET domain-containing lysine methyltransferase 7 (SETD7), a methyltransferase, has been implicated in OA pathology. This study investigates the expression pattern of SETD7 in OA and its role in promoting interleukin-1 beta (IL-1β)-induced chondrocyte injury through modulation of autophagy and inflammation. METHODS The expression of SETD7 in cartilage tissues from OA patients and healthy controls was quantified using quantitative reverse transcription PCR and Western blot analysis. Small interfering RNA targeting SETD7 (si-SETD7) was transfected into human articular chondrocytes (HACs) treated with IL-1β to examine its impact on cellular viability, apoptosis, inflammatory responses, and autophagy. Functional assays including Cell Counting Kit-8, flow cytometry, enzyme-linked immunosorbent assay, and commercial kits were employed to assess biochemical changes. Interaction between YTH N6-methyladenosine RNA binding protein 2 (YTHDF2) and SETD7 was explored using RNA immunoprecipitation and co-immunoprecipitation assays. RESULTS SETD7 was overexpressed in OA cartilage compared with controls and increased further upon IL-1β treatment. Knockdown of SETD7 in IL-1β-treated HACs improved cellular viability, decreased apoptosis, and reversed the adverse effects on lactate dehydrogenase release and inflammatory markers (tumor necrosis factor-alpha and interleukin-6) while enhancing antioxidant enzymes (catalase, malondialdehyde, and superoxide dismutase). Additionally, autophagy was restored, as evidenced by changes in the levels of autophagy related 5, Beclin1, and sequestosome 1. Interfering with autophagy using chloroquine negated the protective effects of SETD7 knockdown. Furthermore, YTHDF2 was found to stabilize SETD7 mRNA, influencing its expression and enhancing IL-1β-induced chondrocyte injury. CONCLUSION SETD7 plays a critical role in the pathogenesis of OA by modulating chondrocyte survival, apoptosis, inflammation, and autophagy. The interaction between YTHDF2 and SETD7 exacerbates chondrocyte injury under inflammatory conditions, highlighting potential therapeutic targets for OA treatment. The YTHDF2/SETD7 axis offers a novel insight into the molecular mechanisms governing cartilage degeneration in OA.
Collapse
Affiliation(s)
- Lexiang Li
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Jun Zhu
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Yi Chen
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Haobo Li
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Yaguang Han
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Lei Zhang
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| | - Bo Wang
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital (The Second Affiliated Hospital of Naval Medical University), Shanghai, China
| |
Collapse
|
16
|
Wahl L, Samson Chillon T, Seemann P, Ohrndorf S, Ochwadt R, Becker W, Schomburg L, Hoff P. Serum selenium, selenoprotein P and glutathione peroxidase 3 in rheumatoid, psoriatic, juvenile idiopathic arthritis, and osteoarthritis. J Nutr Biochem 2025; 135:109776. [PMID: 39389271 DOI: 10.1016/j.jnutbio.2024.109776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/20/2024] [Accepted: 09/28/2024] [Indexed: 10/12/2024]
Abstract
Selenoprotein P (SELENOP) controls selenium (Se) transport, and glutathione peroxidase 3 (GPx3) elicits antioxidant activity in blood. Inflammation associates with Se deficiency, but knowledge concerning selenoproteins in inflammatory rheumatic musculoskeletal diseases (iRMD) is limited. We compared three Se biomarkers in patients with rheumatoid (RA), psoriatic (PsA), and juvenile idiopathic arthritis (JIA) in comparison to osteoarthritis (OA) and healthy subjects, to improve the data base on selenoprotein expression in iRMD. The cross-sectional study enrolled n=272 patients with RA (n=131), PsA (n=67), JIA (n=22) and OA (n=52). Serum Se was quantified by total reflection X-ray fluorescence, SELENOP by ELISA and GPx3 by an enzymatic test. Data from the EPIC trial served as reference. Impairment of daily life was assessed by the Functional Ability Questionnaire (FfbH). Serum SELENOP and Se concentrations correlated linearly in all groups and were below the average measured in EPIC. Se concentration was not different between the patient groups. Compared to controls, SELENOP levels were low in iRMD patients. GPx3 activity was particularly low in JIA and PsA. Seropositive but not seronegative RA patients displayed a disrupted interaction between GPx3 and Se or SELENOP. SELENOP associated with the functional status measured by the FfbH, most pronounced in OA (R=0.76, P < .01). The data indicate selenoprotein deficiency in the majority of patients with iRMD, and a positive relation of SELENOP with functional status in OA. Since increased Se supply improves selenoprotein biosynthesis, a personalized correction of diagnosed deficiency merits consideration to improve Se transport and ameliorate disease burden.
Collapse
Affiliation(s)
- Lukas Wahl
- MVZ Endokrinologikum Berlin am Gendarmenmarkt, Berlin, Germany; Charité Universitätsmedizin Berlin, Klinik für Rheumatologie und Klinische Immunologie, Berlin, Germany; Charité Universitätsmedizin Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Thilo Samson Chillon
- Charité Universitätsmedizin Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | | | - Sarah Ohrndorf
- Charité Universitätsmedizin Berlin, Klinik für Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Ragna Ochwadt
- MVZ für Laboratoriumsmedizin, Genetik und Mikrobiologie Hamburg GmbH, Hamburg, Germany
| | | | - Lutz Schomburg
- Charité Universitätsmedizin Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany.
| | - Paula Hoff
- MVZ Endokrinologikum Berlin am Gendarmenmarkt, Berlin, Germany; Charité Universitätsmedizin Berlin, Klinik für Rheumatologie und Klinische Immunologie, Berlin, Germany.
| |
Collapse
|
17
|
Zhuang H, Ren X, Li H, Zhang Y, Zhou P. Cartilage-targeting peptide-modified cerium oxide nanoparticles alleviate oxidative stress and cartilage damage in osteoarthritis. J Nanobiotechnology 2024; 22:784. [PMID: 39702137 DOI: 10.1186/s12951-024-03068-1] [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: 03/03/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease that leads to a substantial decline in the well-being of older individuals. Chondrocyte senescence and the resultant damage to cartilage tissue, induced by elevated levels of reactive oxygen species within the joint cavity, are significant causative factors in OA development. Cerium oxide nanoparticles (CeONPs) present a promising avenue for therapeutic investigation due to their exceptional antioxidant properties. However, the limited effectiveness of drugs in the joint cavity is often attributed to their rapid clearance by synovial fluid. METHODS Polyethylene glycol-packed CeONPs (PEG-CeONPs) were synthesized and subsequently modified with the cartilage-targeting peptide WYRGRLGK (WY-PEG-CeO). The antioxidant free radical activity and the mimetic enzyme activity of PEG-CeONPs and WY-PEG-CeO were detected. The impact of WY-PEG-CeO on chondrocytes oxidative stress, cellular senescence, and extracellular matrix degradation was assessed using in vitro assays. The cartilage targeting and protective effects were explored in animal models. RESULTS WY-PEG-CeO demonstrated significant efficacy in inhibiting oxidative stress, cellular senescence, and extracellular matrix degradation in OA chondrocytes. The underlying mechanism involves the inhibition of the PI3K/AKT and MAPK signaling pathways. Animal models further revealed that WY-PEG-CeO exhibited a prolonged residence time and enhanced penetration efficiency in cartilage tissue, leading to the attenuation of pathological changes in OA. CONCLUSIONS These findings suggest that WY-PEG-CeO exerts therapeutic effects in OA by inhibiting oxidative stress and suppressing the over-activation of PI3K/AKT and MAPK signaling pathways. This investigation served as a fundamental step towards the advancement of CeONPs-based interventions, providing potential strategies for the treatment of OA.
Collapse
Affiliation(s)
- Huangming Zhuang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xunshan Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huajie Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuelong Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Panghu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
18
|
Yu C, Zhao S, Yue S, Chen X, Dong Y. Novel insights into the role of metabolic disorder in osteoarthritis. Front Endocrinol (Lausanne) 2024; 15:1488481. [PMID: 39744183 PMCID: PMC11688211 DOI: 10.3389/fendo.2024.1488481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/29/2024] [Indexed: 01/06/2025] Open
Abstract
Osteoarthritis (OA) is a prevalent condition that affects individuals worldwide and is one of the leading causes of disability. Nevertheless, the underlying pathological mechanisms of OA remain inadequately understood. Current treatments for OA include non-drug therapies, pharmacological interventions, and surgical procedures. These treatments are mainly focused on alleviating clinical manifestations and improving patients' quality of life, but are not effective in limiting the progression of OA. The detailed understanding of the pathogenesis of OA is extremely significant for the development of OA treatment. Metabolic syndrome has become a great challenge for medicine and public health, In recent years, several studies have demonstrated that the metabolic syndrome and its individual components play a crucial role in OA. Consequently, this review summarizes the mechanisms and research progress on how metabolic syndrome and its components affect OA. The aim is to gain a deeper understanding of the pathogenesis of OA and explore effective treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yonghui Dong
- Department of Orthopedics, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| |
Collapse
|
19
|
Madabeni A, Bortoli M, Nogara PA, Ribaudo G, Dalla Tiezza M, Flohé L, Rocha JBT, Orian L. 50 Years of Organoselenium Chemistry, Biochemistry and Reactivity: Mechanistic Understanding, Successful and Controversial Stories. Chemistry 2024; 30:e202403003. [PMID: 39304519 PMCID: PMC11639659 DOI: 10.1002/chem.202403003] [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: 08/09/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/22/2024]
Abstract
In 1973, two major discoveries changed the face of selenium chemistry: the identification of the first mammal selenoenzyme, glutathione peroxidase 1, and the discovery of the synthetic utility of the so-called selenoxide elimination. While the chemical mechanism behind the catalytic activity of glutathione peroxidases appears to be mostly unveiled, little is known about the mechanisms of other selenoproteins and, for some of them, even the function lies in the dark. In chemistry, the capacity of organoselenides of catalyzing hydrogen peroxide activation for the practical manipulation of organic functional groups has been largely explored, and some mechanistic details have been clearly elucidated. As a paradox, despite the long-standing experience in the field, the nature of the active oxidant in various reactions still remains matter of debate. While many successes characterize these fields, the pharmacological use of organoselenides still lacks any true application, and while some organoselenides were found to be non-toxic and safe to use, to date no therapeutically approved use was granted. In this review, some fundamental and chronologically aligned topics spanning organoselenium biochemistry, chemistry and pharmacology are discussed, focusing on the current mechanistic picture describing their activity as either bioactive compounds or catalysts.
Collapse
Affiliation(s)
- Andrea Madabeni
- Dipartimento di Scienze ChimicheUniversità degli Studi di PadovaVia Marzolo 135131PadovaItaly
| | - Marco Bortoli
- Department of Chemistry and Hylleraas Centre for Quantum Molecular SciencesUniversity of OsloOslo0315Norway
| | - Pablo A. Nogara
- Instituto Federal de Educação, Ciência e Tecnologia Sul-rio-grandense (IFSul)Av. Leonel de Moura Brizola, 250196418-400Bagé, RSBrasil
| | - Giovanni Ribaudo
- Dipartimento di Medicina Molecolare e TraslazionaleUniversità degli Studi di BresciaViale Europa 1125123BresciaItaly
| | - Marco Dalla Tiezza
- Dipartimento di Scienze ChimicheUniversità degli Studi di PadovaVia Marzolo 135131PadovaItaly
| | - Leopold Flohé
- Department of Molecular MedicineUniversity of PadovaItaly
- Departamento de BioquímicaUniversidad de la RepúblicaMontevideoUruguay
| | - João B. T. Rocha
- Departamento de BioquímicaUniversidade Federaldo Rio Grande do Sul (UFRGS)90035-003Porto Alegre, RSBrazil
| | - Laura Orian
- Dipartimento di Scienze ChimicheUniversità degli Studi di PadovaVia Marzolo 135131PadovaItaly
| |
Collapse
|
20
|
Yang F, Shu R, Dai W, Li B, Liu C, Yang H, Johnson HM, Yu S, Bai D, Yang W, Deng Y. H 2Se-evolving bio-heterojunctions promote cutaneous regeneration in infected wounds by inhibiting excessive cellular senescence. Biomaterials 2024; 311:122659. [PMID: 38861831 DOI: 10.1016/j.biomaterials.2024.122659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Pathogenic infection leads to excessive senescent cell accumulation and stagnation of wound healing. To address these issues, we devise and develop a hydrogen selenide (H2Se)-evolving bio-heterojunction (bio-HJ) composed of graphene oxide (GO) and FeSe2 to deracinate bacterial infection, suppress cellular senescence and remedy recalcitrant infected wounds. Excited by near-infrared (NIR) laser, the bio-HJ exerts desired photothermal and photodynamic effects, resulting in rapid disinfection. The crafted bio-HJ could also evolve gaseous H2Se to inhibit cellular senescence and dampen inflammation. Mechanism studies reveal the anti-senescence effects of H2Se-evolving bio-HJ are mediated by selenium pathway and glutathione peroxidase 1 (GPX1). More critically, in vivo experiments authenticate that the H2Se-evolving bio-HJ could inhibit cellular senescence and potentiate wound regeneration in rats. As envisioned, our work not only furnishes the novel gasotransmitter-delivering bio-HJ for chronic infected wounds, but also gets insight into the development of anti-senescence biomaterials.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Shu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenyu Dai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chuang Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Hang Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Hannah M Johnson
- Department of Chemistry, Washington State University, Washington, USA
| | - Sheng Yu
- Department of Chemistry, Washington State University, Washington, USA
| | - Ding Bai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weizhong Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China.
| | - Yi Deng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Pang L, Xiang L, Chen G, Cui W. In-situ hydrogen-generating injectable short fibers for osteoarthritis treatment by alleviating oxidative stress. Acta Biomater 2024; 188:406-419. [PMID: 39293567 DOI: 10.1016/j.actbio.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/21/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
Hydrogen (H₂) has great potential in the treatment of osteoarthritis, but its rapid diffusion and short retention time make it difficult to exert stable therapeutic effects. This study developed a short-fiber injectable material that can continuously generate hydrogen in situ to eliminate reactive oxygen species (ROS), alleviate oxidative stress and inflammation, and promote tissue repair. We prepared H-Si nanosheets with high hydrogen generation efficiency using a wet chemical exfoliation method and combined them with GelMA short fibers via electrospinning technology, achieving the in situ delivery of H-Si nanosheets and regulated hydrogen generation rate through the encapsulation and degradation of GelMA, ultimately achieving continuous and controlled hydrogen supply and stable therapeutic effects for osteoarthritis. In vitro and in vivo experiments confirmed the safety and efficacy of this material. The results showed that the material could continuously and efficiently generate hydrogen in simulated physiological environments (100 mg of material could generate 8.6 % hydrogen), effectively eliminate cellular reactive oxygen species (ROS positive rate reduced by 85.89 %), reduce cellular senescence and apoptosis (cell death rate decreased by 52 %, SA-βgal expression decreased by 78.3 %), promote normal chondrocyte function (Col II expression increased by 67.4 %, Ki67 expression increased by 87.5 %), and improve osteoarthritis in rats (OARSI score increased by 216 %). The in situ hydrogen generation and control system designed in this study provides a new method for the hydrogen's local and stable treatment of osteoarthritis. STATEMENT OF SIGNIFICANCE: Hydrogen (H₂) has great potential in the treatment of osteoarthritis by alleviating oxidative stress, but its rapid diffusion and short retention time make it difficult to exert stable therapeutic effects. This study introduces an innovative injectable material combining H-Si nanosheets and GelMA short fibers to address this issue. By enabling continuous in situ hydrogen generation, this material effectively eliminates reactive oxygen species, reduces oxidative stress and inflammation, and promotes tissue repair. In vitro and in vivo experiments demonstrate its high hydrogen generation efficiency, safety, and therapeutic efficacy, offering a promising new approach for osteoarthritis management.
Collapse
Affiliation(s)
- Libin Pang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China; Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, the Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, PR China
| | - Lei Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Gang Chen
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, the Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
22
|
Kieronska-Rudek A, Kij A, Bar A, Kurpinska A, Mohaissen T, Grosicki M, Stojak M, Sternak M, Buczek E, Proniewski B, Kuś K, Suraj-Prazmowska J, Panek A, Pietrowska M, Zapotoczny S, Shanahan CM, Szabo C, Chlopicki S. Phylloquinone improves endothelial function, inhibits cellular senescence, and vascular inflammation. GeroScience 2024; 46:4909-4935. [PMID: 38980631 PMCID: PMC11336140 DOI: 10.1007/s11357-024-01225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/24/2024] [Indexed: 07/10/2024] Open
Abstract
Phylloquinon (PK) and menaquinones (MK) are both naturally occurring compounds belonging to vitamin K group. Present study aimed to comprehensively analyze the influence of PK in several models of vascular dysfunction to determine whether PK has vasoprotective properties, similar to those previously described for MK. Effects of PK and MK on endothelial dysfunction were studied in ApoE/LDLR-/- mice in vivo, in the isolated aorta incubated with TNF, and in vascular cells as regard inflammation and cell senescence (including replicative and stress-induced models of senescence). Moreover, the vascular conversion of exogenous vitamins to endogenous MK-4 was analyzed. PK, as well as MK, given for 8 weeks in diet (10 mg/kg) resulted in comparable improvement in endothelial function in the ApoE/LDLR-/- mice. Similarly, PK and MK prevented TNF-induced impairment of endothelium-dependent vasorelaxation in the isolated aorta. In in vitro studies in endothelial and vascular smooth muscle cells, we identified that both PK and MK displayed anti-senescence effects via decreasing DNA damage while in endothelial cells anti-inflammatory activity was ascribed to the modulation of NFκB activation. The activity of PK and MK was comparable in terms of their effect on senescence and inflammation. Presence of endogenous synthesis of MK-4 from PK in aorta and endothelial and smooth muscle cells suggests a possible involvement of MK in vascular effects of PK. In conclusion, PK and MK display comparable vasoprotective effects, which may be ascribed, at least in part, to the inhibition of cell senescence and inflammation. The vasoprotective effect of PK in the vessel wall can be related to the direct effects of PK, as well as to the action of MK formed from PK in the vascular wall.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marek Grosicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Elżbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Kuś
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish Academy of Sciences, Krakow, Poland
| | - Monika Pietrowska
- Centre for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Szczepan Zapotoczny
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Catherine M Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King's College London, London, UK
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland.
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
23
|
Yang JC, Liu M, Huang RH, Zhao L, Niu QJ, Xu ZJ, Wei JT, Lei XG, Sun LH. Loss of SELENOW aggravates muscle loss with regulation of protein synthesis and the ubiquitin-proteasome system. SCIENCE ADVANCES 2024; 10:eadj4122. [PMID: 39303039 DOI: 10.1126/sciadv.adj4122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/13/2024] [Indexed: 09/22/2024]
Abstract
Sarcopenia is characterized by accelerated muscle mass and function loss, which burdens and challenges public health worldwide. Several studies indicated that selenium deficiency is associated with sarcopenia; however, the specific mechanism remains unclear. Here, we demonstrated that selenoprotein W (SELENOW) containing selenium in the form of selenocysteine functioned in sarcopenia. SELENOW expression is up-regulated in dexamethasone (DEX)-induced muscle atrophy and age-related sarcopenia mouse models. Knockout (KO) of SELENOW profoundly aggravated the process of muscle mass loss in the two mouse models. Mechanistically, SELENOW KO suppressed the RAC1-mTOR cascade by the interaction between SELENOW and RAC1 and induced the imbalance of protein synthesis and degradation. Consistently, overexpression of SELENOW in vivo and in vitro alleviated the muscle and myotube atrophy induced by DEX. SELENOW played a role in age-related sarcopenia and regulated the genes associated with aging. Together, our study uncovered the function of SELENOW in age-related sarcopenia and provides promising evidence for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Jia-Cheng Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Meng Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Rong-Hui Huang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Qin-Jian Niu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ze-Jing Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jin-Tao Wei
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY 14853, USA
| | - Lv-Hui Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal Breeding and Sustainable Production, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
24
|
Qin Z, Li X, Wang P, Liu Q, Li Y, Gu A, Jiang Q, Gu N. Ultrasmall Prussian Blue Nanozyme Attenuates Osteoarthritis by Scavenging Reactive Oxygen Species and Regulating Macrophage Phenotype. NANO LETTERS 2024; 24:11697-11705. [PMID: 39225479 DOI: 10.1021/acs.nanolett.4c03314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by obscure etiology and unsatisfactory therapeutic outcomes, making the development of new efficient therapies urgent. Superfluous reactive oxygen species (ROS) have historically been considered one of the crucial factors inducing the pathological progression of OA. Ultrasmall Prussian blue nanoparticles (USPBNPs), approximately sub-5 nm in size, are developed by regulating the configuration of polyvinylpyrrolidone chains. USPBNPs display an excellent ROS eliminating capacity and catalase-like activity, capable of decomposing hydrogen peroxide (H2O2) into O2. The anti-inflammatory mechanism of USPBNPs can be attributed to repolarizing macrophages from pro-inflammatory M1 to anti-inflammatory M2 phenotype by decreasing the ROS levels accompanied by O2 improvement. Additionally, USPBNPs exhibit an exciting therapeutic efficiency against OA, comparable to that of hydrocortisone in vivo. This study not only develops a new therapeutic agent for OA but also offers an estimable insight into the application of the nanozyme.
Collapse
Affiliation(s)
- Zhiguo Qin
- Department of Pharmacy, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaofei Li
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
- Department of Sport Medicine, The Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical University, Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu 222023, China
| | - Peng Wang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Qian Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yan Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210009, China
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine and Offspring Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
25
|
Geng N, Fan M, Kuang B, Zhang F, Xian M, Deng L, Chen C, Pan Y, Chen J, Feng N, Liang L, Ye Y, Liu K, Li X, Du Y, Guo F. 10-hydroxy-2-decenoic acid prevents osteoarthritis by targeting aspartyl β hydroxylase and inhibiting chondrocyte senescence in male mice preclinically. Nat Commun 2024; 15:7712. [PMID: 39231947 PMCID: PMC11375154 DOI: 10.1038/s41467-024-51746-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/15/2024] [Indexed: 09/06/2024] Open
Abstract
Osteoarthritis is a degenerative joint disease with joint pain as the main symptom, caused by fibrosis and loss of articular cartilage. Due to the complexity and heterogeneity of osteoarthritis, there is a lack of effective individualized disease-modifying osteoarthritis drugs in clinical practice. Chondrocyte senescence is reported to participate in occurrence and progression of osteoarthritis. Here we show that small molecule 10-hydroxy-2-decenoic acid suppresses cartilage degeneration and relieves pain in the chondrocytes, cartilage explants from osteoarthritis patients, surgery-induced medial meniscus destabilization or naturally aged male mice. We further confirm that 10-hydroxy-2-decenoic acid exerts a protective effect by targeting the glycosylation site in the Asp_Arg_Hydrox domain of aspartyl β-hydroxylase. Mechanistically, 10-hydroxy-2-decenoic acid alleviate cellular senescence through the ERK/p53/p21 and GSK3β/p16 pathways in the chondrocytes. Our study uncovers that 10-hydroxy-2-decenoic acid modulate cartilage metabolism by targeting aspartyl β-hydroxylase to inhibit chondrocyte senescence in osteoarthritis. 10-hydroxy-2-decenoic acid may be a promising therapeutic drug against osteoarthritis.
Collapse
Affiliation(s)
- Nana Geng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Mengtian Fan
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Biao Kuang
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengmei Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Menglin Xian
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Lin Deng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Cheng Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiming Pan
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jianqiang Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Naibo Feng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Li Liang
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yuanlan Ye
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Kaiwen Liu
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Xiaoli Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yu Du
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fengjin Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
26
|
Zhou R, Guo J, Jin Z. Advancing osteoarthritis therapy with GMOCS hydrogel-loaded BMSCs-exos. J Nanobiotechnology 2024; 22:493. [PMID: 39160590 PMCID: PMC11334447 DOI: 10.1186/s12951-024-02713-z] [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: 03/05/2024] [Accepted: 07/08/2024] [Indexed: 08/21/2024] Open
Abstract
This study investigated the mechanism of the extracellular matrix-mimicking hydrogel-mediated TGFB1/Nrf2 signaling pathway in osteoarthritis using bone marrow mesenchymal stem cell-derived exosomes (BMSCs-Exos). A GMOCS-Exos hydrogel was synthesized and evaluated for its impact on chondrocyte viability and neutrophil extracellular traps (NETs) formation. In an OA rat model, GMOCS-Exos promoted cartilage regeneration and inhibited NETs formation. Transcriptome sequencing identified TGFB1 as a key gene, with GMOCS-Exos activating Nrf2 signaling through TGFB1. Depletion of TGFB1 hindered the cartilage-protective effect of GMOCS-Exos. This study sheds light on a promising therapeutic strategy for osteoarthritis through GMOCS-Exos-mediated TGFB1/Nrf2 pathway modulation.
Collapse
Affiliation(s)
- Renyi Zhou
- Department of Orthopedics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001, Liaoning Province, China
| | - Jiarong Guo
- Department of Orthopedics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001, Liaoning Province, China
| | - Zhe Jin
- Department of Orthopedics, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
27
|
Ahmed Mohamed Z, Yang J, Wen J, Jia F, Banerjee S. SEPHS1 Gene: A new master key for neurodevelopmental disorders. Clin Chim Acta 2024; 562:119844. [PMID: 38960024 DOI: 10.1016/j.cca.2024.119844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
The SEPHS1 (Selenophosphate Synthetase 1) gene encodes a critical enzyme for synthesizing selenophosphate, the active donor of selenium (Se) necessary for selenoprotein biosynthesis. Selenoproteins are vital for antioxidant defense, thyroid hormone metabolism, and cellular homeostasis. Mutations in SEPHS1 gene, are associated with neurodevelopmental disorders with developmental delay, poor growth, hypotonia, and dysmorphic features. Due to Se's critical role in brain development and function, SEPHS1 gene has taken center stage in neurodevelopmental research. This review explores the structure and function of the SEPHS1 gene, its role in neurodevelopment, and the implications of its dysregulation for neurodevelopmental disorders. Therapeutic strategies, including Se supplementation, gene therapy, and targeted therapies, are discussed as potential interventions to address SEPHS1 associated neurodevelopmental dysfunction. The study's findings reveal how SEPHS1 mutations disrupt neurodevelopment, emphasizing the gene's intolerance to loss of function. Future research should focus on functional characterization of SEPHS1 variants, broader genetic screenings, and therapeutic developments.
Collapse
Affiliation(s)
- Zakaria Ahmed Mohamed
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jianli Yang
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Jianping Wen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Feiyong Jia
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Santasree Banerjee
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
28
|
Lee H, Choe J, Son MH, Lee IH, Lim MJ, Jeon J, Yang S. A Novel BD2-Selective Inhibitor of BRDs Mitigates ROS Production and OA Pathogenesis. Antioxidants (Basel) 2024; 13:943. [PMID: 39199189 PMCID: PMC11352053 DOI: 10.3390/antiox13080943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Bromodomain and extra-terminal domain (BET) family proteins regulate transcription and recognize lysine residues in histones. Selective BET inhibitors targeting one domain have attracted attention because they maintain normal physiological activities, whereas pan (nonselective) BET inhibitors do not. Osteoarthritis (OA) is a joint disorder characterized by cartilage degeneration for which no treatment currently exists. Here, we investigated whether the selective inhibition of BET proteins is an appropriate therapeutic strategy for OA. We focused on the development and characterization of 2-(4-(2-(dimethylamino)ethoxy)-3,5-dimethylphenyl)-5,7-dimethoxyquinazolin-4(3H)-one (BBC0906), a novel bromodomain 2 (BD2)-specific inhibitor designed to suppress OA progression. Using a DNA-encoded chemical library (DEL) screening approach, BBC0906 was identified because of its high affinity with the BD2 domain of BET proteins. BBC0906 effectively reduced reactive oxygen species (ROS) production and suppressed catabolic factor expression in chondrocytes in vitro. Moreover, in an OA mouse model induced by the destabilization of the medial meniscus (DMM), BBC0906 intra-articular injection attenuated cartilage degradation and alleviated OA. Importantly, BBC0906 selectively inhibits the BD2 domain, thus minimizing its potential side effects. We highlighted the therapeutic potential of targeting BET proteins to modulate oxidative stress and suppress cartilage degradation in OA. BBC0906 is a promising candidate for OA treatment, offering improved safety and efficacy.
Collapse
Affiliation(s)
- Hyemi Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Jihye Choe
- Benobio Co., Ltd., Seongnam-si 13494, Republic of Korea; (J.C.); (M.-H.S.); (I.-H.L.)
| | - Min-Hee Son
- Benobio Co., Ltd., Seongnam-si 13494, Republic of Korea; (J.C.); (M.-H.S.); (I.-H.L.)
| | - In-Hyun Lee
- Benobio Co., Ltd., Seongnam-si 13494, Republic of Korea; (J.C.); (M.-H.S.); (I.-H.L.)
| | - Min Ju Lim
- Department of Biomedical Sciences, Graduate School of Medicine, Ajou University, Suwon 16499, Republic of Korea;
| | - Jimin Jeon
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| |
Collapse
|
29
|
Lee H, Nam J, Jang H, Park YS, Son MH, Lee IH, Eyun SI, Yang JH, Jeon J, Yang S. BRD2-specific inhibitor, BBC0403, inhibits the progression of osteoarthritis pathogenesis in osteoarthritis-induced C57BL/6 male mice. Br J Pharmacol 2024; 181:2528-2544. [PMID: 38600628 DOI: 10.1111/bph.16359] [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: 06/25/2023] [Revised: 01/10/2024] [Accepted: 02/20/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND AND PURPOSE The discovery of new bromo- and extra-terminal inhibitors presents new drugs to treat osteoarthritis (OA). EXPERIMENTAL APPROACH The new drug, BBC0403, was identified in the DNA-encoded library screening system by searching for compounds that target BRD (bromodomain-containing) proteins. The binding force with BRD proteins was evaluated using time-resolved fluorescence energy transfer (TR-FRET) and binding kinetics assays. Subsequently, in vitro and ex vivo analyses demonstrated the effects of the BRD2 inhibitor, BBC0403, on OA. For animal experiments, medial meniscus destabilization was performed to create a 12-week-old male C57BL/6 mouse model, and intra-articular (i.a.) injections were administered. Histological and immunohistochemical analyses were then performed. The underlying mechanism was confirmed by gene set enrichment analysis (GSEA) using RNA-seq. KEY RESULTS TR-FRET and binding kinetics assays revealed that BBC0403 exhibited higher binding specificity for BRD2 compared to BRD3 and BRD4. The anti-OA effects of BBC0403 were tested at concentrations of 5, 10 and 20 μM (no cell toxicity in the range tested). The expression of catabolic factors, prostaglandin E2 (PGE2) production and extracellular matrix (ECM) degradation was reduced. Additionally, the i.a. injection of BBC0403 prevented OA cartilage degradation in mice. Finally, BBC0403 was demonstrated to suppress NF-κB and MAPK signalling pathways. CONCLUSION AND IMPLICATIONS This study demonstrated that BBC0403 is a novel BRD2-specific inhibitor and a potential i.a.-injectable therapeutic agent to treat OA.
Collapse
Affiliation(s)
- Hyemi Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - Jiho Nam
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - Hahyeong Jang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | | | | | | | - Seong-Il Eyun
- Department of Life Science, Chung-Ang University, Seoul, South Korea
| | - Jae-Hyun Yang
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute Harvard Medical School (HMS), Boston, Massachusetts, USA
| | - Jimin Jeon
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
30
|
Zou B, Xiong Z, Yu Y, Shi S, Li X, Chen T. Rapid Selenoprotein Activation by Selenium Nanoparticles to Suppresses Osteoclastogenesis and Pathological Bone Loss. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401620. [PMID: 38621414 DOI: 10.1002/adma.202401620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/28/2024] [Indexed: 04/17/2024]
Abstract
Osteoclast hyperactivation stands as a significant pathological factor contributing to the emergence of bone disorders driven by heightened oxidative stress levels. The modulation of the redox balance to scavenge reactive oxygen species emerges as a viable approach to addressing this concern. Selenoproteins, characterized by selenocysteine (SeCys2) as the active center, are crucial for selenium-based antioxidative stress therapy for inflammatory diseases. This study reveals that surface-active elemental selenium (Se) nanoparticles, particularly lentinan-Se (LNT-Se), exhibit enhanced cellular accumulation and accelerated metabolism to SeCys2, the primary active Se form in biological systems. Consequently, LNT-Se demonstrates significant inhibition of osteoclastogenesis. Furthermore, in vivo studies underscore the superior therapeutic efficacy of LNT-Se over SeCys2, potentially attributable to the enhanced stability and safety profile of LNT-Se. Specifically, LNT-Se effectively modulates the expression of the selenoprotein GPx1, thereby exerting regulatory control over osteoclastogenesis inhibition, and the prevention of osteolysis. In summary, these results suggest that the prompt activation of selenoproteins by Se nanoparticles serves to suppress osteoclastogenesis and pathological bone loss by upregulating GPx1. Moreover, the utilization of bioactive Se species presents a promising avenue for effectively managing bone disorders.
Collapse
Affiliation(s)
- Binhua Zou
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Zushuang Xiong
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yanzi Yu
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Sujiang Shi
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Department of Chemistry, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Laboratory of Viral Pathogenesis & Infection Prevention and Control of Ministry of Education, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
31
|
Geng N, Xian M, Deng L, Kuang B, Pan Y, Liu K, Ye Y, Fan M, Bai Z, Guo F. Targeting the senescence-related genes MAPK12 and FOS to alleviate osteoarthritis. J Orthop Translat 2024; 47:50-62. [PMID: 39007035 PMCID: PMC11245888 DOI: 10.1016/j.jot.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/07/2024] [Accepted: 06/03/2024] [Indexed: 07/16/2024] Open
Abstract
Background The mechanism by which chondrocyte senescence aggravate OA progression has not yet been well elucidated. The aim of this study was to investigate the chondrocyte senescence related gene biosignatures in OA, and to analyze on the underlying mechanisms of senescence in OA. Materials and methods We intersected osteoarthritis dataset GSE82107 from GEO database and senescence dataset from CellAge database of human senescence-associated genes based on genetic manipulations experiments plus gene expression profilin, and screened out 4 overlapping genes. The hub genes were verified in vitro and in human OA cartilage tissues by qRT-PCR. We further confirmed the function of mitogen-activated protein kinase 12 (MAPK12) and Fos proto-oncogene (FOS) in OA in vitro and in vivo by qRT-PCR, western blotting, Edu staining, immunofluorescence, SA-β-gal staining, HE, IHC, von frey test, and hot plate. Results 1458 downregulated and 218 upregulated DEGs were determined from GSE82107, and 279 human senescence-associated genes were downloaded from CellAge database. After intersection assay, we screened out 4 overlapping genes, of which FOS, CYR61 and TNFSF15 were upregulated, MAPK12 was downregulated. The expression of MAPK12 was obviously downregulated, whereas the expression profiles of FOS, CYR61 and TNFSF15 were remarkedly upregulated in H2O2- or IL-1β-stimulated C28/I2 cells, human OA cartilage tissues, and knee cartilage of aging mice. Furthermore, both MAPK12 over-expression and FOS knock-down can promote cell proliferation and cartilage anabolism, inhibit cell senescence and cartilage catabolism, relieve joint pain in H2O2- or IL-1β-stimulated C28/I2 cells and mouse primary chondrocytes, destabilization of the medial meniscus (DMM) mice. Conclusion This study explored that MAPK12 and FOS are involved in the occurrence and development of OA through modulating chondrocyte senescence. They might be biomarkers of OA chondrocyte senescence, and provides some evidence as subsequent possible therapeutic targets for OA. The translational potential of this article The translation potential of this article is that we revealed MAPK12 and FOS can effectively alleviate OA by regulating chondrocyte senescence, and thus provided potential therapeutic targets for prevention or treatment of OA in the future.
Collapse
Affiliation(s)
- Nana Geng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Menglin Xian
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Lin Deng
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Biao Kuang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiming Pan
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Kaiwen Liu
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yuanlan Ye
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Mengtian Fan
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Zhixun Bai
- Department of Nephrology, The First Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Fengjin Guo
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Basic Medical Sciences, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
Cho Y, Kim H, Yook G, Yong S, Kim S, Lee N, Kim YJ, Kim JH, Kim TW, Chang MJ, Lee KM, Chang CB, Kang SB, Kim JH. Predisposal of Interferon Regulatory Factor 1 Deficiency to Accumulate DNA Damage and Promote Osteoarthritis Development in Cartilage. Arthritis Rheumatol 2024; 76:882-893. [PMID: 38268484 DOI: 10.1002/art.42815] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 01/05/2024] [Accepted: 01/23/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVE Interferon regulatory factor 1 (IRF1) is a transcriptional regulator conventionally associated with immunomodulation. Recent molecular analyses mapping DNA binding sites of IRF1 have suggested its potential function in DNA repair. However, the physiologic significance of this noncanonical function remains unexplored. Here, we investigated the role of IRF1 in osteoarthritis (OA), a condition marked by senescence and chronic joint inflammation. METHODS OA progression was examined in wild-type and Irf1-/- mice using histologic assessments and microcomputed tomography analysis of whole-joint OA manifestations and behavioral assessments of joint pain. An integrated analysis of assay for transposase-accessible chromatin with sequencing and whole transcriptome data was conducted for the functional assessment of IRF1 in chondrocytes. The role of IRF1 in DNA repair and senescence was investigated by assaying γ-H2AX foci and senescence-associated beta-galactosidase activity. RESULTS Our genome-wide investigation of IRF1 footprinting in chondrocytes revealed its primary occupancies in the promoters of DNA repair genes without noticeable footprint patterns in those of interferon-responsive genes. Chondrocytes lacking IRF1 accumulated irreversible DNA damage under oxidative stress, facilitating their entry into cellular senescence. IRF1 was down-regulated in the cartilage of human and mouse OA. Although IRF1 overexpression did not elicit an inflammatory response in joints or affect OA development, genetic deletion of Irf1 caused enhanced chondrocyte senescence and exacerbated post-traumatic OA in mice. CONCLUSION IRF1 offers DNA damage surveillance in chondrocytes, protecting them from oxidative stress associated with OA risk factors. Our study provides a crucial and cautionary perspective that compromising IRF1 activity renders chondrocytes vulnerable to cellular senescence and promotes OA development.
Collapse
Affiliation(s)
- Yongsik Cho
- Institute for Basic Science and Seoul National University, Seoul, South Korea, and Liflex Science, Cheongju, South Korea
| | - Hyeonkyeong Kim
- Institute for Basic Science and Seoul National University, Seoul, South Korea, and Liflex Science, Cheongju, South Korea
| | - Geunho Yook
- Institute for Basic Science and Seoul National University, Seoul, South Korea
| | - Sangmin Yong
- Institute for Basic Science and Seoul National University, Seoul, South Korea
| | - Soy Kim
- Institute for Basic Science and Seoul National University, Seoul, South Korea
| | - Narae Lee
- Institute for Basic Science and Seoul National University, Seoul, South Korea
| | - Yi-Jun Kim
- Ewha Womans University, Seoul, South Korea
| | | | - Tae Woo Kim
- Seoul National University and Boramae Hospital, Seoul, South Korea
| | - Moon Jong Chang
- Seoul National University and Boramae Hospital, Seoul, South Korea
| | - Kyoung Min Lee
- Seoul National University and Boramae Hospital, Seoul, South Korea
| | - Chong Bum Chang
- Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seung-Baik Kang
- Seoul National University and Boramae Hospital, Seoul, South Korea
| | - Jin-Hong Kim
- Seoul National University and Institute for Basic Science, Seoul, South Korea, and Institute of Green-Bio Science and Technology, Pyeongchang, South Korea
| |
Collapse
|
33
|
Yuan Y, Li Y, Deng Q, Yang J, Zhang J. Selenadiazole-Induced Hela Cell Apoptosis through the Redox Oxygen Species-Mediated JAK2/STAT3 Signaling Pathway. ACS OMEGA 2024; 9:20919-20926. [PMID: 38764630 PMCID: PMC11097172 DOI: 10.1021/acsomega.3c10107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/21/2024] [Accepted: 04/17/2024] [Indexed: 05/21/2024]
Abstract
Cervical cancer is a significant global health concern, and novel therapeutic strategies are continually being sought to combat this disease. In recent years, selenadiazole found latent therapeutic effects on tumors. Herein, investigating the mechanism of selenadiazole in Hela cells holds promise for advancing cervical cancer treatment. Hela cells, a widely utilized model for studying cervical cancer, were treated with selenadiazole, and cell viability was assessed by using the cell counting kit-8 (CCK-8) assay. Changes in mitochondrial membrane potential were evaluated using JC-1 staining, while apoptosis induction was examined using AnnexinV-PI double staining. Intracellular ROS levels were measured by using specific fluorescent probes and the ELIASA system. Additionally, Western blotting was performed to assess the activation of related proteins in response to selenadiazole. Data analysis was performed using GraphPad. Exposure to selenadiazole led to a substantial increase in intracellular redox oxygen species (ROS) levels in Hela cells. Importantly, the induction of ROS by selenadiazole was associated with a marked increase in mitochondrial apoptosis, as evidenced by elevated levels of AnnexinV-positive cells, the JC-1 monomer, caspase-9, and Bcl-2. Furthermore, activation of the JAK2/STAT3 pathway was observed following the selenadiazole treatment. Selenadiazole holds the potential to suppress tumor growth in cervical cancer cells by increasing reactive oxygen species (ROS) levels and inducing mitochondrial apoptosis via the JAK2/STAT3 pathway. This study offers valuable insights into potential cervical cancer therapies and underscores the need for further research into the specific mechanisms of selenadiazole.
Collapse
Affiliation(s)
- Yi Yuan
- Center
Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Yinghua Li
- Center
Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510120, China
| | - Qinglin Deng
- Nanfang
Hospital, Southern Medical University, Guangzhou 510120, China
| | - Jinying Yang
- Department
of Obstetrics, Longgang District Maternity
and Child Healthcare Hospital of Shenzhen City (Longgang Maternity
and Child Clinical Institute of Shantou University Medical College), Shenzhen 510080, China
| | - Jing Zhang
- Department
of Interventional Radiology, Guangdong Provincial People’s
Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
34
|
Mullegama SV, Kiernan KA, Torti E, Pavlovsky E, Tilton N, Sekula A, Gao H, Alaimo JT, Engleman K, Rush ET, Blocker K, Dipple KM, Fettig VM, Hare H, Glass I, Grange DK, Griffin M, Phornphutkul C, Massingham L, Mehta L, Miller DE, Thies J, Merritt JL, Muller E, Osmond M, Sawyer SL, Slaugh R, Hickey RE, Wolf B, Choudhary S, Simonović M, Zhang Y, Palculict TB, Telegrafi A, Carere DA, Wentzensen IM, Morrow MM, Monaghan KG, Yang J, Juusola J. De novo missense variants in exon 9 of SEPHS1 cause a neurodevelopmental condition with developmental delay, poor growth, hypotonia, and dysmorphic features. Am J Hum Genet 2024; 111:778-790. [PMID: 38531365 PMCID: PMC11023921 DOI: 10.1016/j.ajhg.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Selenophosphate synthetase (SEPHS) plays an essential role in selenium metabolism. Two mammalian SEPHS paralogues, SEPHS1 and SEPHS2, share high sequence identity and structural homology with SEPHS. Here, we report nine individuals from eight families with developmental delay, growth and feeding problems, hypotonia, and dysmorphic features, all with heterozygous missense variants in SEPHS1. Eight of these individuals had a recurrent variant at amino acid position 371 of SEPHS1 (p.Arg371Trp, p.Arg371Gln, and p.Arg371Gly); seven of these variants were known to be de novo. Structural modeling and biochemical assays were used to understand the effect of these variants on SEPHS1 function. We found that a variant at residue Trp352 results in local structural changes of the C-terminal region of SEPHS1 that decrease the overall thermal stability of the enzyme. In contrast, variants of a solvent-exposed residue Arg371 do not impact enzyme stability and folding but could modulate direct protein-protein interactions of SEPSH1 with cellular factors in promoting cell proliferation and development. In neuronal SH-SY5Y cells, we assessed the impact of SEPHS1 variants on cell proliferation and ROS production and investigated the mRNA expression levels of genes encoding stress-related selenoproteins. Our findings provided evidence that the identified SEPHS1 variants enhance cell proliferation by modulating ROS homeostasis. Our study supports the hypothesis that SEPHS1 plays a critical role during human development and provides a basis for further investigation into the molecular mechanisms employed by SEPHS1. Furthermore, our data suggest that variants in SEPHS1 are associated with a neurodevelopmental disorder.
Collapse
Affiliation(s)
- Sureni V Mullegama
- GeneDx, Gaithersburg, MD 20877, USA; Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA.
| | - Kaitlyn A Kiernan
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | - Ethan Pavlovsky
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Nicholas Tilton
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Austin Sekula
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Hua Gao
- GeneDx, Gaithersburg, MD 20877, USA
| | - Joseph T Alaimo
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, MO, USA; Department of Pediatrics, University of Missouri Kansas City, School of Medicine, Kansas City, MO, USA; Center for Pediatric Genomic Medicine, Children's Mercy Hospital, Kansas City, MO, USA
| | - Kendra Engleman
- Department of Pediatrics, University of Missouri Kansas City, School of Medicine, Kansas City, MO, USA; Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Eric T Rush
- Department of Pediatrics, University of Missouri Kansas City, School of Medicine, Kansas City, MO, USA; Division of Clinical Genetics, Children's Mercy Hospital, Kansas City, MO, USA; Department of Internal Medicine, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Karli Blocker
- Division of Clinical Genetics, Stanford Children's Health, San Francisco, CA, USA
| | - Katrina M Dipple
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Veronica M Fettig
- Center for Inherited Cardiovascular Disease, Cardiovascular Genetics Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Heather Hare
- Northeastern Ontario Medical Genetics Program, Health Sciences, North Sudbury, ON, Canada
| | - Ian Glass
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Dorothy K Grange
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Griffin
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Chanika Phornphutkul
- Division of Genetics, Department of Pediatrics, Alpert School of Medicine at Brown University, Providence, RI, USA
| | - Lauren Massingham
- Division of Genetics, Department of Pediatrics, Alpert School of Medicine at Brown University, Providence, RI, USA
| | - Lakshmi Mehta
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Danny E Miller
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Jenny Thies
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA, USA
| | - J Lawrence Merritt
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Eric Muller
- Division of Clinical Genetics, Stanford Children's Health, San Francisco, CA, USA
| | - Matthew Osmond
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Sarah L Sawyer
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Rachel Slaugh
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel E Hickey
- Department of Pediatrics, Division of Genetics, Birth Defects and Metabolism, Anne & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Barry Wolf
- Department of Pediatrics, Division of Genetics, Birth Defects and Metabolism, Anne & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Sanjeev Choudhary
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Miljan Simonović
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | | | | | | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | | |
Collapse
|
35
|
Chen L, Shao Z, Zhang Z, Teng W, Mou H, Jin X, Wei S, Wang Z, Eloy Y, Zhang W, Zhou H, Yao M, Zhao S, Chai X, Wang F, Xu K, Xu J, Ye Z. An On-Demand Collaborative Innate-Adaptive Immune Response to Infection Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304774. [PMID: 37523329 DOI: 10.1002/adma.202304774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Deep tissue infection is a common clinical issue and therapeutic difficulty caused by the disruption of the host antibacterial immune function, resulting in treatment failure and infection relapse. Intracellular pathogens are refractory to elimination and can manipulate host cell biology even after appropriate treatment, resulting in a locoregional immunosuppressive state that leads to an inadequate response to conventional anti-infective therapies. Here, a novel antibacterial strategy involving autogenous immunity using a biomimetic nanoparticle (NP)-based regulating system is reported to induce in situ collaborative innate-adaptive immune responses. It is observed that a macrophage membrane coating facilitates NP enrichment at the infection site, followed by active NP accumulation in macrophages in a mannose-dependent manner. These NP-armed macrophages exhibit considerably improved innate capabilities, including more efficient intracellular ROS generation and pro-inflammatory factor secretion, M1 phenotype promotion, and effective eradication of invasive bacteria. Furthermore, the reprogrammed macrophages direct T cell activation at infectious sites, resulting in a robust adaptive antimicrobial immune response to ultimately achieve bacterial clearance and prevent infection relapse. Overall, these results provide a conceptual framework for a novel macrophage-based strategy for infection treatment via the regulation of autogenous immunity.
Collapse
Affiliation(s)
- Liang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Zhenxuan Shao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Zengjie Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Wangsiyuan Teng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Xiaoqiang Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Shenyu Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
| | - Zenan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Yinwang Eloy
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Hao Zhou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Minjun Yao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Shenzhi Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Xupeng Chai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Orthopedics Research Institute of Zhejiang University, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, 310000, P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou City, 310000, P. R. China
| |
Collapse
|
36
|
Zhang D, Zhang Y, Xia S, Shen P, Yang C. Metabolic profiling of synovial fluid in human temporomandibular joint osteoarthritis. Front Immunol 2024; 15:1335181. [PMID: 38529278 PMCID: PMC10961395 DOI: 10.3389/fimmu.2024.1335181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024] Open
Abstract
Introduction Temporomandibular joint (TMJ) osteoarthritis (OA) is a common TMJ degenerative disease with an unclear mechanism. Synovial fluid (SF), an important component of TMJ, contains various proteins and metabolites that may directly contribute to OA. The present study aimed to investigate the influence of SF in TMJOA at the metabolite level. Methods Untargeted and widely targeted metabolic profiling were employed to identify metabolic changes in SF of 90 patients with different TMJOA grades according to TMJ magnetic resonance imaging. Results A total 1498 metabolites were detected. Most of the metabolites were amino acids and associated metabolites, benzene and substituted derivatives, and lipids. Among patients with mild, moderate and severe TMJOA, 164 gradually increasing and 176 gradually decreasing metabolites were identified, indicating that biosynthesis of cofactors, choline metabolism, mineral absorption and selenocompound metabolism are closely related to TMJOA grade. Combined metabolomics and clinical examination revealed 37 upregulated metabolites and 16 downregulated metabolites in patients with pain, of which 19 and 26 metabolites were positively and negatively correlated, respectively, with maximum interincisal opening. A model was constructed to diagnose TMJOA grade and nine biomarkers were identified. The identified metabolites are key to exploring the mechanism of TMJOA. Discussion In the present study, a metabolic profile was constructed and assessed using a much larger number of human SF samples from patients with TMJOA, and a model was established to contribute to the diagnosis of TMJOA grade. The findings expand our knowledge of metabolites in human SF of TMJOA patients, and provide an important basis for further research on the pathogenesis and treatment of TMJOA.
Collapse
Affiliation(s)
- Dahe Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yuxin Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Simo Xia
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Pei Shen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
37
|
Zhang DG, Kunz WS, Lei XJ, Zito E, Zhao T, Xu YC, Wei XL, Lv WH, Luo Z. Selenium Ameliorated Oxidized Fish Oil-Induced Lipotoxicity via the Inhibition of Mitochondrial Oxidative Stress, Remodeling of Usp4-Mediated Deubiquitination, and Stabilization of Pparα. Antioxid Redox Signal 2024; 40:433-452. [PMID: 37265154 DOI: 10.1089/ars.2022.0194] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Aims: Studies demonstrated that oxidized fish oil (OFO) promoted oxidative stress and induced mitochondrial dysfunction and lipotoxicity, which attenuated beneficial effects of fish oil supplements in the treatment of nonalcoholic fatty liver disease (NAFLD). The current study was performed on yellow catfish, a good model to study NAFLD, and its hepatocytes to explore whether selenium (Se) could alleviate OFO-induced lipotoxicity via the inhibition of oxidative stress and determine its potential mechanism. Results: The analysis of triglycerides content, oxidative stress parameters, and histological and transmission electronic microscopy observation showed that high dietary Se supplementation alleviated OFO-induced lipotoxicity, oxidative stress, and mitochondrial injury and dysfunction. RNA-sequencing and immunoblotting analysis indicated that high dietary Se reduced OFO-induced decline of peroxisome-proliferator-activated receptor alpha (Pparα) and ubiquitin-specific protease 4 (Usp4) protein expression. High Se supplementation also alleviated OFO-induced reduction of thioredoxin reductase 2 (txnrd2) messenger RNA (mRNA) expression level and activity. The txnrd2 knockdown experiments revealed that txnrd2 mediated Se- and oxidized eicosapentaenoic acid (oxEPA)-induced changes of mitochondrial reactive oxygen species (mtROS) and further altered Usp4 mediated-deubiquitination and stabilization of Pparα, which, in turn, modulated mitochondrial fatty acid β-oxidation and metabolism. Mechanistically, Usp4 deubiquitinated Pparα and ubiquitin-proteasome-mediated Pparα degradation contributed to oxidative stress-induced mitochondrial dysfunction. Innovation: These findings uncovered a previously unknown mechanism by which Se and OFO interacted to affect lipid metabolism via the Txnrd2-mtROS-Usp4-Pparα pathway, which provides the new target for NAFLD prevention and treatment. Conclusion: Se ameliorated OFO-induced lipotoxicity via the inhibition of mitochondrial oxidative stress, remodeling of Usp4-mediated deubiquitination, and stabilization of Pparα. Antioxid. Redox Signal. 40, 433-452.
Collapse
Affiliation(s)
- Dian-Guang Zhang
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Wolfram S Kunz
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
- Department of Epileptology, University of Bonn, Bonn, Germany
| | - Xi-Jun Lei
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Tao Zhao
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Yi-Chuang Xu
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Xiao-Lei Wei
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Wu-Hong Lv
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Zhi Luo
- Shenzhen Institute of Nutrition and Health, Fishery College, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
38
|
Lan S, Huang H, Liu W, Xu C, Lei X, Dong W, Liu J, Yang S, Cotman AE, Zhang Q, Fang X. Asymmetric Transfer Hydrogenation of Cyclobutenediones. J Am Chem Soc 2024; 146:4942-4957. [PMID: 38326715 DOI: 10.1021/jacs.3c14239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Four-membered carbocycles are fundamental substructures in bioactive molecules and approved drugs and serve as irreplaceable building blocks in organic synthesis. However, developing efficient protocols furnishing diversified four-membered ring compounds in a highly regio-, diastereo-, and enantioselective fashion remains challenging but very desirable. Here, we report the unprecedented asymmetric transfer hydrogenation of cyclobutenediones. The reaction can selectively afford three types of four-membered products in high yields with high stereoselectivities, and the highly functionalized products enable a series of further transformations to form more diversified four-membered compounds. Asymmetric synthesis of di-, tri-, and tetrasubstituted bioactive molecules has also been achieved. Systematic mechanistic studies and theoretical calculations have revealed the origin of the regioselectivity, the key hydrogenation transition state models, and the sequence of the double and triple hydrogenation processes. The work provides a new choice for the catalytic asymmetric synthesis of cyclobutanes and related structures and demonstrates the robustness of asymmetric transfer hydrogenation in the accurate selectivity control of highly functionalized substrates.
Collapse
Affiliation(s)
- Shouang Lan
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| | - Huangjiang Huang
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
- Fujian Normal University, Fuzhou 350108, China
| | - Wenjun Liu
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| | - Chao Xu
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| | - Xiang Lei
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| | - Wennan Dong
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| | - Jinggong Liu
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510120, China
| | - Shuang Yang
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| | - Andrej Emanuel Cotman
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Qi Zhang
- School of Chemistry and Chemical Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xinqiang Fang
- State Key Laboratory of Structural Chemistry, and Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Center for Excellence in Molecular Synthesis, Fujian Institute of Research on the Structure of Matter, Fujian College, University of Chinese Academy of Sciences, Fuzhou 350100, China
| |
Collapse
|
39
|
Dang JY, Zhang W, Chu Y, Chen JH, Ji ZL, Feng P. Downregulation of salusins alleviates hypertrophic cardiomyopathy via attenuating oxidative stress and autophagy. Eur J Med Res 2024; 29:109. [PMID: 38336819 PMCID: PMC10854150 DOI: 10.1186/s40001-024-01676-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/15/2024] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Salusins, which are translated from the alternatively spliced mRNA of torsin family 2 member A (TOR2A), play a vital role in regulation of various cardiovascular diseases. However, it remains unclear precisely regarding their roles in hypertrophic cardiomyopathy (HCM). Therefore, this study was conducted to explore therapeutic effect and the underlying mechanisms of salusins on HCM. MATERIAL AND METHODS In vivo experiments, Sprague-Dawley rats were used to induce HCM model by angiotensin (Ang) II infusion for 4 weeks. The rats were randomly divided into four groups, namely, Saline + Control shRNA (n = 7), Ang II + Control shRNA (n = 8), Saline + TOR2A shRNA (n = 7), and Ang II + TOR2A shRNA groups (n = 8). After HCM induction, doppler echocardiography is recommended to evaluate heart function. In vitro experiments, primary neonatal rat cardiomyocytes (NRCMs) and cardiac fibroblasts (NRCFs) were obtained from newborn rats, and were treated with Ang II (10-6 M) for 24 h. RESULTS After treatment with Ang II, levels of salusin-α and salusin-β were elevated in serum and cardiac tissues of rats and in the neonatal rat cardiomyocytes and cardiac fibroblasts. Downregulation of salusins alleviated the Ang II-induced cardiac hypertrophy by suppressing the increased atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and beta-myosin heavy chain (β-MHC) and cardiac fibrosis by blocking collagen I, collagen III and transforming growth factor-beta (TGF-β), and it also attenuated oxidative stress by suppressing the increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels and reversing the decreased superoxide dismutase (SOD) activity and autophagy by inhibiting the increased microtubule-associated protein light chain 3B (LC3B), Beclin1, autophagy related gene (Atg) 3 and Atg5 in the cardiac tissues of Ang II-infused rats and in the Ang II-treated NRCMs. CONCLUSIONS All these findings suggest that the levels of salusins were elevated in the HCM, and targeting of salusins contributes to alleviation of cardiac hypertrophy and fibrosis probably via attenuating oxidative stress and autophagy. Accordingly, targeting of salusins may be a strategy for HCM therapy.
Collapse
Affiliation(s)
- Jing-Yi Dang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Wei Zhang
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Yi Chu
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Jiang-Hong Chen
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Zhao-Le Ji
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China
| | - Pin Feng
- Department of Cardiology, Tangdu Hospital, Airforce Medical University, No. 569 Xinsid Road, Xi'an, 710038, China.
| |
Collapse
|
40
|
Hu T, Shi Z, Sun Y, Hu F, Rong Y, Wang J, Wang L, Xu W, Zhang F, Zhang WZ. SEPHS1 attenuates intervertebral disc degeneration by delaying nucleus pulposus cell senescence through the Hippo-Yap/Taz pathway. Am J Physiol Cell Physiol 2024; 326:C386-C399. [PMID: 38105759 DOI: 10.1152/ajpcell.00571.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Nucleus pulposus cell (NPC) senescence is a major cause of intervertebral disc degeneration (IVDD). Oxidative stress and reactive oxygen species (ROS) play critical roles in regulating cell senescence. Selenophosphate synthetase 1 (SEPHS1) was reported to play an important role in mitigating oxidative stress in an osteoarthritis (OA) model by reducing the production of ROS, thereby, delaying the occurrence and development of osteoarthritis. In this study, we explored the, hitherto unknown, role of SEPHS1 in IVDD in vitro and in vivo using an interleukin-1β (IL-1β)-induced NPC senescence model and a rat needle puncture IVDD model, respectively. SEPHS1 delayed NPC senescence in vitro by reducing ROS production. Age-related dysfunction was also ameliorated by the overexpression of SEPHS1 and inhibition of the Hippo-Yap/Taz signaling pathway. In vivo experiments revealed that the overexpression of SEPHS1 and inhibition of Hippo-Yap/Taz alleviated IVDD in rats. Moreover, a selenium (Se)-deficient diet and lack of SEPHS1 synergistically aggravated IVDD progression. Taken together, our results demonstrate that SEPHS1 plays a significant role in NPC senescence. Overexpression of SEPHS1 and inhibition of Hippo-Yap/Taz can delay NPC senescence, restore the balance of extracellular matrix metabolism, and attenuate IVDD. SEPHS1 could be a promising therapeutic target for IVDD.NEW & NOTEWORTHY Selenophosphate synthetase 1 (SEPHS1) deficiency leads to an increase in reactive oxygen species levels and in the subsequent activation of the Hippo-Yap/Taz signaling pathway. In the rat model of intervertebral disc degeneration (IVDD), overexpression of SEPHS1 and inhibition of Hippo-YAP/Taz mitigated the progression of disc degeneration indicating the involvement of SEPHS1 in IVDD. SEPHS1 is a promising therapeutic target for IVDD.
Collapse
Affiliation(s)
- Tao Hu
- Department of Orthopedics, Provincial Hospital Affiliated to Anhui Medical University, Hefei, People's Republic of China
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Zhongming Shi
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yongjin Sun
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Feng Hu
- Department of Orthopedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China
| | - Yuluo Rong
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Jia Wang
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Liang Wang
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Wenbin Xu
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Feng Zhang
- Division of Life Sciences and Medicine, Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, People's Republic of China
| | - Wen-Zhi Zhang
- Department of Orthopedics, Provincial Hospital Affiliated to Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
41
|
Fu L, Duan H, Cai Y, Chen X, Zou B, Yuan L, Liu G. Moxibustion ameliorates osteoarthritis by regulating gut microbiota via impacting cAMP-related signaling pathway. Biomed Pharmacother 2024; 170:116031. [PMID: 38113621 DOI: 10.1016/j.biopha.2023.116031] [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: 08/22/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent progressive disorder. Moxibustion has found widespread use in clinical practice for OA, while its underlying mechanism remains elusive. OBJECTIVE To investigate whether moxibustion can ameliorate OA by influencing the metabolic processes in OA and to elucidate the specific metabolic mechanisms involved. METHODS C57BL/6J WT mice were randomly assigned to one of three groups: the SHAM group, the ACLT group, and the ACLT+M group. In the ACLT+M group, mice underwent moxibustion treatment at acupoints Shenshu (BL23) and Zusanli (ST36) for a continuous period of 28 days, with each session lasting 20 min. We conducted a comprehensive analysis to assess the impact of moxibustion on OA, focusing on pathological changes, intestinal flora composition, and serum metabolites. RESULTS Moxibustion treatment effectively mitigated OA-related pathological changes. Specifically, moxibustion treatment resulted in the amelioration of articular cartilage damage, synovial inflammation, subchondral bone sclerosis when compared to the ACLT group. Moreover, 16S rDNA sequencing analysis revealed that moxibustion treatment positively influenced the composition of the flora, making it more similar to that of the SHAM group. Notably, moxibustion treatment led to a reduction in the abundance of Ruminococcus and Proteobacteria in the intestine. In addition, non-targeted metabolomics analysis identified 254 significantly different metabolites between the groups. Based on KEGG pathway analysis and the observed impact of moxibustion on OA-related inflammation, moxibustion therapy is closely associated with the cAMP-related signaling pathway. CONCLUSION Moxibustion can relieve OA by regulating intestinal flora and via impacting cAMP-related signaling pathway.
Collapse
Affiliation(s)
- Liping Fu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Huimin Duan
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yisi Cai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xuelan Chen
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Binhua Zou
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Lixia Yuan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
42
|
Qi Z, Duan A, Ng K. Selenoproteins in Health. Molecules 2023; 29:136. [PMID: 38202719 PMCID: PMC10779588 DOI: 10.3390/molecules29010136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Selenium (Se) is a naturally occurring essential micronutrient that is required for human health. The existing form of Se includes inorganic and organic. In contrast to the inorganic Se, which has low bioavailability and high cytotoxicity, organic Se exhibits higher bioavailability, lower toxicity, and has a more diverse composition and structure. This review presents the nutritional benefits of Se by listing and linking selenoprotein (SeP) functions to evidence of health benefits. The research status of SeP from foods in recent years is introduced systematically, particularly the sources, biochemical transformation and speciation, and the bioactivities. These aspects are elaborated with references for further research and utilization of organic Se compounds in the field of health.
Collapse
Affiliation(s)
- Ziqi Qi
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Alex Duan
- Melbourne TrACEES Platform, School of Chemistry, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Ken Ng
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia;
| |
Collapse
|
43
|
Wei G, Lu K, Umar M, Zhu Z, Lu WW, Speakman JR, Chen Y, Tong L, Chen D. Risk of metabolic abnormalities in osteoarthritis: a new perspective to understand its pathological mechanisms. Bone Res 2023; 11:63. [PMID: 38052778 PMCID: PMC10698167 DOI: 10.1038/s41413-023-00301-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/27/2023] [Indexed: 12/07/2023] Open
Abstract
Although aging has traditionally been viewed as the most important risk factor for osteoarthritis (OA), an increasing amount of epidemiological evidence has highlighted the association between metabolic abnormalities and OA, particularly in younger individuals. Metabolic abnormalities, such as obesity and type II diabetes, are strongly linked to OA, and they affect both weight-bearing and non-weight-bearing joints, thus suggesting that the pathogenesis of OA is more complicated than the mechanical stress induced by overweight. This review aims to explore the recent advances in research on the relationship between metabolic abnormalities and OA risk, including the impact of abnormal glucose and lipid metabolism, the potential pathogenesis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Guizheng Wei
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ke Lu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Muhammad Umar
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhenglin Zhu
- Department of Orthopedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - William W Lu
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - John R Speakman
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yan Chen
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
44
|
Chen Y, Zhang X, Yang J, Feng W, Deng G, Xu S, Guo M. Extracellular Vesicles Derived from Selenium-Deficient MAC-T Cells Aggravated Inflammation and Apoptosis by Triggering the Endoplasmic Reticulum (ER) Stress/PI3K-AKT-mTOR Pathway in Bovine Mammary Epithelial Cells. Antioxidants (Basel) 2023; 12:2077. [PMID: 38136197 PMCID: PMC10740620 DOI: 10.3390/antiox12122077] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/28/2023] [Accepted: 11/04/2023] [Indexed: 12/24/2023] Open
Abstract
Selenium (Se) deficiency disrupts intracellular REDOX homeostasis and severely deteriorates immune and anti-inflammatory function in high-yielding periparturient dairy cattle. To investigate the damage of extracellular vesicles derived from Se-deficient MAC-T cells (SeD-EV) on normal mammary epithelial cells, an in vitro model of Se deficiency was established. Se-deficient MAC-T cells produced many ROS, promoting apoptosis and the release of inflammatory factors. Extracellular vesicles were successfully isolated by ultrahigh-speed centrifugation and identified by transmission electron microscopy, particle size analysis, and surface markers (CD63, CD81, HSP70, and TSG101). RNA sequencing was performed on exosomal RNA. A total of 9393 lncRNAs and 63,155 mRNAs transcripts were identified in the SeC and SeD groups, respectively, of which 126 lncRNAs and 955 mRNAs were differentially expressed. Furthermore, SeD-EV promoted apoptosis of normal MAC-T cells by TUNEL analysis. SeD-EV significantly inhibited Bcl-2, while Bax and Cleaved Caspase3 were greatly increased. Antioxidant capacity (CAT, T-AOC, SOD, and GSH-Px) was inhibited in SeD-EV-treated MAC-T cells. Additionally, p-PERK, p-eIF2α, ATF4, CHOP, and XBP1 were all elevated in MAC-T cells supplemented with SeD-EV. In addition, p-PI3K, p-Akt, and p-mTOR were decreased strikingly by SeD-EV. In conclusion, SeD-EV caused oxidative stress, thus triggering apoptosis and inflammation through endoplasmic reticulum stress and the PI3K-Akt-mTOR signaling pathway, which contributed to explaining the mechanism of Se deficiency causing mastitis.
Collapse
Affiliation(s)
- Yu Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.C.); (S.X.)
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Xiangqian Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Z.); (J.Y.); (W.F.); (G.D.)
| | - Jing Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Z.); (J.Y.); (W.F.); (G.D.)
| | - Wen Feng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Z.); (J.Y.); (W.F.); (G.D.)
| | - Ganzhen Deng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (X.Z.); (J.Y.); (W.F.); (G.D.)
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.C.); (S.X.)
| | - Mengyao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.C.); (S.X.)
| |
Collapse
|
45
|
Kong K, Jin M, Zhao C, Qiao H, Chen X, Li B, Rong K, Zhang P, Shan Y, Xu Z, Chang Y, Li H, Zhai Z. Mechanical overloading leads to chondrocyte degeneration and senescence via Zmpste24-mediated nuclear membrane instability. iScience 2023; 26:108119. [PMID: 37965144 PMCID: PMC10641493 DOI: 10.1016/j.isci.2023.108119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/28/2023] [Accepted: 09/29/2023] [Indexed: 11/16/2023] Open
Abstract
Patients with OA and varus knees are subject to abnormal mechanical environment and objective of this study was to investigate the molecular mechanisms underlying chondrocyte senescence caused by mechanical overloading and the role of Zmpste24-mediated nuclear membrane instability in varus knees. Finite element analysis showed that anteromedial region of tibial plateau experienced the most mechanical stress in an osteoarthritis patient with a varus knee. Immunohistochemistry exhibited lower Zmpste24 expression and higher expression of senescence marker p21 in the anteromedial region. Animal experiments and cell-stretch models also demonstrated an inverse relationship between Zmpste24 and mechanically induced senescence. Zmpste24 overexpression rescued cartilage degeneration and senescence in vitro by scavenging ROS. In conclusion, anteromedial tibial plateau is exposed to abnormal stress in varus knees, downregulation of Zmpste24, and nuclear membrane stability may explain increased senescence in this region. Zmpste24 and nuclear membrane stability are potential targets for treating osteoarthritis caused by abnormal alignment.
Collapse
Affiliation(s)
- Keyu Kong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minghao Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chen Zhao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuzhuo Chen
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, College of Stomatology, Ninth People’s Hospital, Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baixing Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Pu Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Shan
- Suzhou Ninth People’s Hospital, Department of Orthopedics, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou, China
| | - Zhengquan Xu
- Suzhou Municipal Hospital, Department of Orthopedics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Zhang F, Cheng T, Zhang SX. Mechanistic target of rapamycin (mTOR): a potential new therapeutic target for rheumatoid arthritis. Arthritis Res Ther 2023; 25:187. [PMID: 37784141 PMCID: PMC10544394 DOI: 10.1186/s13075-023-03181-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by systemic synovitis and bone destruction. Proinflammatory cytokines activate pathways of immune-mediated inflammation, which aggravates RA. The mechanistic target of rapamycin (mTOR) signaling pathway associated with RA connects immune and metabolic signals, which regulates immune cell proliferation and differentiation, macrophage polarization and migration, antigen presentation, and synovial cell activation. Therefore, therapy strategies targeting mTOR have become an important direction of current RA treatment research. In the current review, we summarize the biological functions of mTOR, its regulatory effects on inflammation, and the curative effects of mTOR inhibitors in RA, thus providing references for the development of RA therapeutic targets and new drugs.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Xinghualing District, Taiyuan, 030001, Shanxi Province, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Ting Cheng
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Xinghualing District, Taiyuan, 030001, Shanxi Province, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Xinghualing District, Taiyuan, 030001, Shanxi Province, China.
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi Province, China.
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China.
| |
Collapse
|
47
|
Lee H, Nam J, Jang H, Park YS, Son MH, Lee IH, Eyun SI, Jeon J, Yang S. Novel molecule BBC0901 inhibits BRD4 and acts as a catabolic regulator in the pathogenesis of osteoarthritis. Biomed Pharmacother 2023; 166:115426. [PMID: 37666177 DOI: 10.1016/j.biopha.2023.115426] [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: 07/10/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023] Open
Abstract
Osteoarthritis (OA) is induced by matrix degradation and inflammation mediated by bromo-domain-containing protein 4 (BRD4)-dependent catabolic factors. BRD4 acts as both a transcriptional regulator and an epigenetic reader. BBC0901 was identified as an inhibitor of BRD4 using a DNA-encoded library screening system. We aimed to demonstrate the effects of BBC0901 on OA pathogenesis by in vitro, ex vivo, and in vivo analyses. BBC0901 inhibited the expression of catabolic factors that degrade cartilage without significantly affecting the viability of mouse articular chondrocytes. Additionally, ex vivo experiments under conditions mimicking OA showed that BBC0901 suppressed extracellular matrix degradation. RNA sequencing analysis of gene expression patterns showed that BBC0901 inhibited the expression of catabolic factors, such as matrix metalloproteinases (MMPs) and cyclooxygenase (COX)2, along with reactive oxygen species (ROS) production. Furthermore, intra-articular (IA) injection of BBC0901 into the knee joint blocked osteoarthritic cartilage destruction by inhibition of MMP3, MMP13, COX2, interleukin (IL)6, and ROS production, thereby obstructing the nuclear factor kappa-light-chain-enhancer of activated B cell and mitogen activated protein kinase signaling. In conclusion, BBC0901-mediated BRD4 inhibition prevented OA development by attenuating catabolic signaling and hence, can be considered a promising IA therapeutic for OA.
Collapse
Affiliation(s)
- Hyemi Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jiho Nam
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Hahyeong Jang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Young-Sik Park
- Benobio Co., Ltd., Gyeonggi-do 13494, Republic of Korea.
| | - Min-Hee Son
- Benobio Co., Ltd., Gyeonggi-do 13494, Republic of Korea.
| | - In-Hyun Lee
- Benobio Co., Ltd., Gyeonggi-do 13494, Republic of Korea.
| | - Seong-Il Eyun
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Jimin Jeon
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Siyoung Yang
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
48
|
Xu C, Xia Y, Zhuang P, Liu W, Mu C, Liu Z, Wang J, Chen L, Dai H, Luo Z. FePSe 3 -Nanosheets-Integrated Cryogenic-3D-Printed Multifunctional Calcium Phosphate Scaffolds for Synergistic Therapy of Osteosarcoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303636. [PMID: 37217971 DOI: 10.1002/smll.202303636] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/11/2023] [Indexed: 05/24/2023]
Abstract
Clinical treatment of osteosarcoma encounters great challenges of postsurgical tumor recurrence and extensive bone defect. To develop an advanced artificial bone substitute that can achieve synergistic bone regeneration and tumor therapy for osteosarcoma treatment, a multifunctional calcium phosphate composite enabled by incorporation of bioactive FePSe3 -nanosheets within the cryogenic-3D-printed α-tricalcium phosphate scaffold (TCP-FePSe3 ) is explored. The TCP-FePSe3 scaffold exhibits remarkable tumor ablation ability due to the excellent NIR-II (1064 nm) photothermal property of FePSe3 -nanosheets. Moreover, the biodegradable TCP-FePSe3 scaffold can release selenium element to suppress tumor recurrence by activating of the caspase-dependent apoptosis pathway. In a subcutaneous tumor model, it is demonstrated that tumors can be efficiently eradicated via the combination treatment with local photothermal ablation and the antitumor effect of selenium element. Meanwhile, in a rat calvarial bone defect model, the superior angiogenesis and osteogenesis induced by TCP-FePSe3 scaffold have been observed in vivo. The TCP-FePSe3 scaffold possesses improved capability to promote the repair of bone defects via vascularized bone regeneration, which is induced by the bioactive ions of Fe, Ca, and P released during the biodegradation of the implanted scaffolds. The TCP-FePSe3 composite scaffolds fabricated by cryogenic-3D-printing illustrate a distinctive strategy to construct multifunctional platform for osteosarcoma treatment.
Collapse
Affiliation(s)
- Chao Xu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yuhao Xia
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Pengzhen Zhuang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Wenliang Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Congpu Mu
- Center for High Pressure Science, State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao, 066004, China
| | - Zhongyuan Liu
- Center for High Pressure Science, State Key Laboratory of Metastable Materials Science and Technology, Yanshan University, Qinhuangdao, 066004, China
| | - Jianglin Wang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhiqiang Luo
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
49
|
Mao X, Yan B, Chen H, Lai P, Ma J. BRG1 mediates protective ability of spermidine to ameliorate osteoarthritic cartilage by Nrf2/KEAP1 and STAT3 signaling pathway. Int Immunopharmacol 2023; 122:110593. [PMID: 37423156 DOI: 10.1016/j.intimp.2023.110593] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Spermidine (SPD) is a natural polyamine that shows beneficial effects on osteoarthritis (OA). However, the effect of SPD on cartilage inflammation remains unknown. This study aimed to investigate the potential mechanisms underlying the protective effect of SPD against OA-induced articular cartilage degradation. METHOD SW1353 human chondrocytes were treated with hydrogen peroxide and lipopolysaccharide to induce models of inflammation and oxidative stress, followed by different dose of SPD intervention. Moreover, mice that underwent anterior cruciate ligament transection were bred and treated with SPD. The effects of SPD were observed using a CCK-8 kit, real-time polymerase chain reaction, immunoblotting, and immunofluorescent assays. RESULT SPD significantly increased the expression of antioxidant proteins, chondrogenic genes, and inflammatory factors both in vivo and in vitro. And injury of the mouse cartilage was also reduced by SPD. Moreover, SPD activated the Nrf2/KEAP1 pathway and inhibited STAT3 phosphorylation. BRG1 expression was decreased in osteoarthritic mouse cartilage, whereas SPD treatment caused an upregulation. However, when BRG1 was specifically inhibited by an adeno-associated virus and small interfering RNA, the antioxidant and anti-inflammatory effects of SPD were significantly diminished both in vitro and in vivo. CONCLUSION We found that SPD ameliorated cartilage damage in OA by activating the BRG1-mediated Nrf2/KEAP1 pathway. SPD and BRG1 may provide new therapeutic options or targets for the treatment of OA.
Collapse
Affiliation(s)
- Xinjie Mao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Yan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongjie Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
50
|
Liang Y, Shen L, Ni W, Ding Y, Yang W, Gu T, Zhang C, Yik JHN, Haudenschild DR, Fan S, Shen S, Hu Z. CircGNB1 drives osteoarthritis pathogenesis by inducing oxidative stress in chondrocytes. Clin Transl Med 2023; 13:e1358. [PMID: 37537733 PMCID: PMC10400757 DOI: 10.1002/ctm2.1358] [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: 03/29/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have risen to prominence as important regulators of biological processes. This study investigated whether circGNB1 functions as a competitive endogenous RNA to regulate the pathological process of oxidative stress in age-related osteoarthritis (OA). METHODS The relationship between circGNB1 expression and oxidative stress/OA severity was determined in cartilages from OA patients at different ages. The biological roles of circGNB1 in oxidative stress and OA progression, and its downstream targets were determined using gain- and loss-of-function experiments in various biochemical assays in human chondrocytes (HCs). The in vivo effects of circGNB1 overexpression and knockdown were also determined using a destabilization of the medial meniscus (DMM) mouse model. RESULTS Increased circGNB1 expression was detected in HCs under oxidative and inflammatory stress and in the cartilage of older individuals. Mechanistically, circGNB1 sponged miR-152-3p and thus blocked its interaction with its downstream mRNA target, ring finger protein 219 (RNF219), which in turn stabilized caveolin-1 (CAV1) by preventing its ubiquitination at the K47 residue. CircGNB1 inhibited IL-10 signalling by antagonizing miR-152-3p-mediated RNF219 and CAV1 inhibition. Consequently, circGNB1 overexpression promoted OA progression by enhancing catabolic factor expression and oxidative stress and by suppressing anabolic genes in vitro and in vivo. Furthermore, circGNB1 knockdown alleviated the severity of OA, whereas circGNB1 overexpression had the opposite effect in a DMM mouse model of OA. CONCLUSION CircGNB1 regulated oxidative stress and OA progression via the miR-152-3p/RNF219/CAV1 axis. Modulating circGNB1 could be an effective strategy for treating OA.
Collapse
Affiliation(s)
- Yi Liang
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Lifeng Shen
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Weiyu Ni
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Yuhong Ding
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Wentao Yang
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Tianyuan Gu
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Chenfeng Zhang
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Jasper H. N. Yik
- Ellison Musculoskeletal Research CenterDepartment of Orthopaedic SurgeryUniversity of California SystemDavisCaliforniaUSA
| | - Dominik R. Haudenschild
- Ellison Musculoskeletal Research CenterDepartment of Orthopaedic SurgeryUniversity of California SystemDavisCaliforniaUSA
| | - Shunwu Fan
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Shuying Shen
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| | - Ziang Hu
- Department of Orthopedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang ProvinceHangzhouChina
| |
Collapse
|