1
|
Lei XT, Chen XF, Qiu S, Tang JY, Geng S, Yang GY, Wu QN. TERT/FOXO1 signaling promotes islet β-cell dysfunction in type 2 diabetes mellitus by regulating ATG9A-mediated autophagy. World J Diabetes 2025; 16:102994. [DOI: 10.4239/wjd.v16.i5.102994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/09/2025] [Accepted: 03/19/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a severe global health problem that causes prolonged disease exposure and an elevated risk for chronic complications, posing a substantial health burden. Although therapies, such as GLP-1 receptor agonists and SGLT2 inhibitors, have been successfully developed, new therapeutic options are still expected to offer better blood glucose control and decrease complications.
AIM To elucidate the mechanism by which TERT/FOXO1 affects high glucose (HG)-induced dysfunction in islet β-cells via the regulation of ATG9A-mediated autophagy.
METHODS High-fat diet (HFD)-fed/streptozotocin (STZ)-treated mice or HG-treated MIN6 cells were used to establish T2DM models. Fasting blood glucose (FBG) and insulin levels in mice, as well as morphological changes in islet tissues, were assessed. Cell proliferation and the apoptosis rate were measured via EdU assays and flow cytometry, respectively. The expression levels of TERT, FOXO1, ATG9A and autophagy-related proteins (LC3B, p62) were analyzed via western blotting. The relationship between FOXO1 and ATG9A was assessed using dual-luciferase reporter gene assays and ChIP assays.
RESULTS T2DM modeling in HFD-fed/STZ-treated mice and HG-treated MIN6 cells led to elevated TERT and FOXO1 expression and reduced ATG9A expression. Mice with T2DM were found to have decreased body weight, worsened morphology, elevated FBG and suppressed insulin levels. HG-treated MIN6 cells presented decreased viability and LC3B expression, in addition to increased p62 expression and apoptosis rates. FOXO1 knockdown both in vitro and in vivo protected mice and cells against islet β-cell dysfunction via the activation of autophagy. The molecular mechanism involved the suppression of ATG9A expression by TERT through FOXO1 transcription activation.
CONCLUSION Our results suggested that TERT/FOXO1 inhibits ATG9A expression to decrease islet β-cell function in T2DM.
Collapse
Affiliation(s)
- Xiao-Tian Lei
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 402360, China
| | - Xiang-Fen Chen
- Department of Endocrinology, The Affiliated Dazu’s Hospital of Chongqing Medical University, The People's Hospital of Dazu, Chongqing 402360, China
| | - Sheng Qiu
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 402360, China
| | - Jia-Ying Tang
- Department of Endocrinology, The Affiliated Dazu’s Hospital of Chongqing Medical University, The People's Hospital of Dazu, Chongqing 402360, China
| | - Shan Geng
- Department of Endocrinology, The Affiliated Dazu’s Hospital of Chongqing Medical University, The People's Hospital of Dazu, Chongqing 402360, China
| | - Gang-Yi Yang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 402360, China
| | - Qi-Nan Wu
- Department of Endocrinology, The Affiliated Dazu’s Hospital of Chongqing Medical University, The People's Hospital of Dazu, Chongqing 402360, China
| |
Collapse
|
2
|
Su D, He J, Yuan W, Duan R, Du B, Shi Y, Liu L, Li X. Effect and potential mechanism of modified citrus pectin in 3D printing-based cartilage tissue engineering. Int J Biol Macromol 2025; 311:144022. [PMID: 40345290 DOI: 10.1016/j.ijbiomac.2025.144022] [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: 09/23/2024] [Revised: 04/20/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Modified citrus pectin (MCP) is widely used as a dietary supplement in the food and pharmaceutical industries with pleiotropic bioactivities. Particularly, MCP has effects on chondroprotection and phenotype maintenance of chondrocyte. Here, after confirming its chondroprotective effect in a partial-thickness articular cartilage (AC) defect, the distributions of MCP in cartilage and chondrocytes were investigated using fluorescence labeled-MCP. Then the potential of it in cartilage tissue engineering (CTE) was studied using 3D-printed scaffolds of hybrid hydrogel (GelMA/HAMA/MCP) of MCP, methacryloydylated-gelatin (GelMA) and hyaluronic acid (HAMA). Finally, the mechanism of the scaffolds on chondrogenesis were analyzed through transcriptome sequencing. It was found that MCP could penetrate cartilage, enter chondrocytes and accumulate in lysosome by binding with Gal-3. MCP could promote the proliferation and maintain the phenotype of chondrocytes in continuous passage culture. Mechanistically, MCP-based scaffold could upregulate the expression of genes of chondrogenic markers and growth factors, downregulate genes related to inflammation or degeneration mediators, and modulate autophagy pathways to maintain homeostasis of chondrocytes. Ultimately, MCP-based scaffolds could support chondrocytes adhesion, proliferation, ECM deposition, and enhance the chondrogenesis of the engineered cartilage. Together, our results demonstrate that MCP has great potential in three-dimensional bioprinting (3DBP)-based CTE to enhance the cartilage regeneration.
Collapse
Affiliation(s)
- Danning Su
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Jiayue He
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Wenlong Yuan
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Ruiping Duan
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Bo Du
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Yanping Shi
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin, 300384, PR China
| | - Lingrong Liu
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China
| | - Xuemin Li
- The Key Laboratory of Biomedical Material of Tianjin, State Key Laboratory of Advanced Medical Materials and Devices, Biomedical Barriers Research Center, Chinese Academy of Medical Sciences & Peking Union Medical College Institute of Biomedical Engineering, Tianjin 300192, PR China.
| |
Collapse
|
3
|
Zhang J, Yuan L, Zhang Y, Jin H, Zhao Y, Zeng X, Zou Y, Wang K, Nie X. Loss of SCRG1 in chondrocytes inhibits osteoarthritis by promoting autophagy activity in the temporomandibular joint through inhibition of neurokine receptors. J Oral Facial Pain Headache 2025; 39:196-203. [PMID: 40129438 PMCID: PMC11934739 DOI: 10.22514/jofph.2025.020] [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: 10/10/2024] [Accepted: 01/16/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND To investigate in vitro how scrapie responsive gene 1 (SCRG1) contributes to the development of temporomandibular joint osteoarthritis (TMJOA). METHODS Western blotting was used to identify protein expression. Proinflammatory cytokine levels were assessed by means of an enzyme-linked immunosorbent test. In order to find out whether chondrocytes expressed protein light chain 3B (LC3B), immunofluorescence was utilized. RESULTS In the TMJOA in vitro model, hydrogen peroxide (H2O2) treatment increased the expression of SCRG1, stimulated chondrocyte catabolism and inflammatory response, and blocked autophagy. In chondrocytes, SCRG1 silencing reduces the inflammatory response, catabolism, and autophagy inhibition brought on by H2O2. Concurrently, H2O2 induction triggers the nuclear factor (NF)-κB pathway and nerve growth factor receptor (NGFR). When SCRG1 is downregulated, NGFR expression is inhibited and the NF-κB pathway is blocked. CONCLUSIONS By inhibiting NGFR and blocking the NF-κB pathway, knocking down SCRG1 can prevent H2O2-induced inflammatory response, metabolic breakdown and autophagy inhibition in chondrocytes.
Collapse
Affiliation(s)
- JiaJun Zhang
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - LePing Yuan
- Department of Oral and Maxillofacial
Surgery, The Third People’s Hospital of
Yuhang District, 311115 Hangzhou,
Zhejiang, China
| | - YanYan Zhang
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - HaoYang Jin
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - YeKe Zhao
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - XiaoKe Zeng
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - YanHui Zou
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - KeYu Wang
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| | - Xin Nie
- Department of Oral and Maxillofacial
Surgery, Hospital of Stomatology
Wenzhou Medical University, 325027
Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Liu Y, Song C, Gao S, Zhou D, Lv J, Zhou Y, Wang L, Shi H, Liu F, Xiong Z, Hou Y, Liu Z. Chondrocyte Ferritinophagy as a Molecular Mechanism of Arthritis-A Narrative Review. Cell Biochem Biophys 2025; 83:1021-1033. [PMID: 39306824 DOI: 10.1007/s12013-024-01534-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] [Accepted: 09/07/2024] [Indexed: 03/03/2025]
Abstract
Osteoarthritis (OA) is a prevalent joint disease affecting orthopedic patients. Its incidence is steadily increasing, causing great economic hardship for individuals and society as a whole. OA is connected with risk factors such as genetics, obesity, and joint diseases; yet, its pathophysiology is still largely understood. At present, several cell death pathways govern the initiation and advancement of OA. It has been discovered that the onset and progression of OA are strongly associated with pyroptosis, senescence, apoptosis, ferroptosis, and autophagy. Ferroptosis and autophagy have not been well studied in OA, and elucidating their molecular mechanisms in chondrocytes is important for the diagnosis of OA. For this reason, we aim was reviewed recent national and international developments and provided an initial understanding of the molecular pathways underlying autophagy and ferroptosis in OA. We determined the reference period to be the last five years by searching for the keywords "osteoarthritis, mechanical stress, Pizeo1, ferroptosis, autophagy, ferritin autophagy" in the three databases of PUBMED, Web of Science, Google Scholar. We then screened irrelevant literature by reading the abstracts. Ferroptosis is a type of programmed cell death that is dependent on reactive oxygen species and Fe2+. It is primarily caused by processes linked to amino acid metabolism, lipid peroxidation, and iron metabolism. Furthermore, Piezoelectric mechanically sensitive ion channel assembly 1 (PIEZO1), which is triggered by mechanical stress, has been revealed to be intimately associated with ferroptosis events. It was found that mechanical injury triggers changes in the intracellular environment of articular chondrocytes (e.g., elevated levels of oxidative stress and increased inflammation) through PIEZO1, ultimately leading to iron death in chondrocytes. Therefore, we believe that PIEZO1 is a key initiator protein of iron death in chondrocytes. Widely present in eukaryotic cells, autophagy is a lysosome-dependent, evolutionarily conserved catabolic process that carries misfolded proteins, damaged organelles, and other macromolecules to lysosomes for breakdown and recycling. Throughout OA, autophagy is activated to differing degrees, indicating that autophagy may play a role in the development of OA. According to recent research, autophagy is a major factor in the process that leads cells to ferroptosis. Despite the notion of ferritinophagy being put forth, not much research has been done to clarify the connection between ferroptosis and autophagy in OA.
Collapse
Affiliation(s)
- Yong Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Chao Song
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Silong Gao
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Daqian Zhou
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jiale Lv
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yang Zhou
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Liquan Wang
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Houyin Shi
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Fei Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China.
| | - Zhongwei Xiong
- Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China.
| | - Yunqing Hou
- Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China.
| | - Zongchao Liu
- Department of Orthopedics and Traumatology (Trauma and Bone-setting), The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Luzhou Longmatan District People's Hospital, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
5
|
Gheyas RN, Williams RC, Ryan KA, Menko AS. The link of FOXO1 and FOXO4 transcription factors to development of the lens. Dev Dyn 2025. [PMID: 39797725 DOI: 10.1002/dvdy.766] [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: 09/21/2024] [Revised: 11/02/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND The FOXOs regulate the transcription of many genes, including ones directly linked to pathways required for lens development. However, this transcription factor family has rarely been studied in the context of development, including the development of the lens. FOXO expression, regulation, and function during lens development remained unexplored. RESULTS In studies of the embryonic lens, we showed that both FOXO1 and FOXO4, which share many downstream targets, are expressed in a differentiation-state-specific manner, most highly in lens epithelial and differentiating cortical fiber cells. Their expression patterns and subcellular distributions suggest both shared and distinct functions. Stabilization of FOXO cytoplasmic pools involved their binding to the chaperone protein 14-3-3. FOXO association with β-catenin linked this transcription complex to fiber cell-specific gene activation. Inhibition of PI3K/Akt signaling promoted FOXO1/FOXO4 nuclear localization in lens epithelial and fiber cells and expression of the CDKi p27 in the lens epithelium where it has been linked to lens cell withdrawal from the cell cycle and initiation of the lens differentiation program. We showed that FOXO1 transcriptional activation is required for the induction of p27 when Akt signaling is blocked, demonstrating the linearity of the PI3K/Akt/FOXO1/p27 pathway. CONCLUSIONS PI3K/Akt signaling regulates FOXO-dependent lens cell differentiation.
Collapse
Affiliation(s)
- Rifah N Gheyas
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ruby C Williams
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kelly A Ryan
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - A Sue Menko
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Hridayanka KSN, Duttaroy AK, Basak S. Bioactive Compounds and Their Chondroprotective Effects for Osteoarthritis Amelioration: A Focus on Nanotherapeutic Strategies, Epigenetic Modifications, and Gut Microbiota. Nutrients 2024; 16:3587. [PMID: 39519419 PMCID: PMC11547880 DOI: 10.3390/nu16213587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
In degenerative joint disease like osteoarthritis (OA), bioactive compounds like resveratrol, epigallocatechin gallate, curcumin, and other polyphenols often target various signalling pathways, including NFκB, TGFβ, and Wnt/β-catenin by executing epigenetic-modifying activities. Epigenetic modulation can target genes of disease pathophysiology via histone modification, promoter DNA methylation, and non-coding RNA expression, some of which are directly involved in OA but have been less explored. OA patients often seek options that can improve the quality of their life in addition to existing treatment with nonsteroidal anti-inflammatory drugs (NSAIDs). Although bioactive and natural compounds exhibit therapeutic potential against OA, several disadvantages loom, like insolubility and poor bioavailability. Nanoformulated bioactive compounds promise a better way to alleviate OA since they also control systemic events, including metabolic, immunological, and inflammatory responses, by modulating host gut microbiota that can regulate OA pathogenesis. Recent data suggest gut dysbiosis in OA. However, limited evidence is available on the role of bioactive compounds as epigenetic and gut modulators in ameliorating OA. Moreover, it is not known whether the effects of polyphenolic bioactive compounds on gut microbial response are mediated by epigenetic modulatory activities in OA. This narrative review highlights the nanotherapeutic strategies utilizing bioactive compounds, reporting their effects on chondrocyte growth, metabolism, and epigenetic modifications in osteoarthritis amelioration.
Collapse
Affiliation(s)
- Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway;
| | - Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| |
Collapse
|
7
|
Zhao C, Feng Y, Zhou Y, Li N, Zhao L. Artesunate attenuates osteoarthritis in mice by promoting MTA1 transcription through a USP7/FoxO1 axis. Toxicol Appl Pharmacol 2024; 491:117075. [PMID: 39173720 DOI: 10.1016/j.taap.2024.117075] [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: 06/24/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Artesunate (ART) is a derivative of artemisinin and has anti-inflammatory, anti-tumor, and anti-angiogenic properties. Although ART has been implicated in osteoarthritis (OA), the mechanism needs to be further dissected. Here, we explored the effects of ART on the development of OA and the underlying mechanism using destabilization of the medial meniscus (DMM) surgical instability model. Mice with OA were developed using DMM and treated with ART. The pathological morphology of knee joint tissues was examined, and the degeneration of joint cartilage was assessed. Mouse knee chondrocytes were isolated and induced with IL-1β, followed by ART treatment. ART alleviates OA in mice by elevating ubiquitin carboxyl-terminal hydrolase 7 (USP7) expression, and USP7 inhibitor (P22077) treatment mitigated the protective effects of ART on chondrocytes. We also showed that USP7 mediated the deubiquitination of forkhead box protein O1 (FoxO1), while FoxO1 alleviated chondrocyte injury. In addition, FoxO1 promoted metastasis-associated protein MTA1 (MTA1) transcription, and downregulation of MTA1 exacerbated chondrocyte injury. Our study identifies that USP7/FoxO1/MTA1 is a key signaling cascade in the treatment of ART on OA.
Collapse
Affiliation(s)
- Chengjin Zhao
- Department of Orthopedics, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, PR China
| | - Yangyang Feng
- Department of Orthopedics, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, PR China
| | - Yuhu Zhou
- Department of Orthopedics, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, PR China
| | - Nannan Li
- Department of Orthopedics, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, PR China
| | - Li Zhao
- Department of Cardiology, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, PR China.
| |
Collapse
|
8
|
Chen Y, Su D, Zheng J, He J, Du B, Duan R, Liu L, Li X. Intra-articular injection of modified citrus pectin and hyaluronate gel induces synergistic effects in treating osteoarthritis. Int J Biol Macromol 2024; 276:133840. [PMID: 39004250 DOI: 10.1016/j.ijbiomac.2024.133840] [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: 09/15/2023] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
We previously found that modified citrus pectin (MCP), an inhibitor of pro-inflammatory factor Galectin-3 (Gal-3), has significant anti-inflammatory and chondroprotective effects. In this study, a hyaluronate (HA) gel-based sustained release system of MCP (MCP-HA) was developed as an anti-inflammatory agent for chronic inflammation for osteoarthritis (OA) treatment. The MCP-HA gel was injected into the knee joint cavities of OA rabbit models induced by anterior cruciate ligament transection (ACLT) or modified Hulth method once a week for five weeks. We found that MCP-HA could improve the symptoms and signs of OA, protect articular cartilage from degeneration, suppress synovial inflammation, and therefore alleviate OA progression. Proteomic analysis of the synovial fluid obtained from the knee joints of OA rabbits revealed that MCP-HA synergistically regulated the levels of multiple inflammatory mediators and proteins involved in metabolic pathways. Taken together, our results demonstrate that the MCP-HA shows a synergistic effect of HA and MCP by modulating both inflammation and metabolic processes, thereby alleviating OA progression. The MCP-HA sustained release system has promising potential for long-term use in OA treatment.
Collapse
Affiliation(s)
- Yazhen Chen
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China
| | - Danning Su
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China
| | - Jianuo Zheng
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China
| | - Jiayue He
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China
| | - Bo Du
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China
| | - Ruiping Duan
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China
| | - Lingrong Liu
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China.
| | - Xuemin Li
- The Key Laboratory of Biomedical Material of Tianjin, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin, 300192, PR, China.
| |
Collapse
|
9
|
Hao Y, Tian X, Yan F, Wang X, Huang J, Li L. Effect of MEHP on testosterone synthesis via Sirt1/Foxo1/Rab7 signaling pathway inhibition of lipophagy in TM3 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116394. [PMID: 38663197 DOI: 10.1016/j.ecoenv.2024.116394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/14/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Mono-2-ethylhexyl phthalic acid (MEHP) is the most toxic metabolite of the plasticizer di-2-ethylhexyl phthalic acid (DEHP), and studies have shown that MEHP causes serious reproductive effects. However, its exact mechanisms of action remain elusive. In this study, we aimed to investigate the reproductive effects of MEHP and preliminarily explore its underlying molecular mechanisms. We found that TM3 cells gradually secreted less testosterone and intracellular free cholesterol with increasing MEHP exposure. MEHP exposure inhibited lipophagy and the Sirt1/Foxo1/Rab7 signaling pathway in TM3 cells, causing aberrant accumulation of intracellular lipid droplets. Addition of the Sirt1 agonist SRT1720 and Rab7 agonist ML-098 alleviated the inhibition of lipophagy and increased free cholesterol and testosterone contents in TM3 cells. SRT1720 alleviated the inhibitory effect of MEHP on the Sirt1/Foxo1/Rab7 signaling pathway, whereas ML-098 only alleviated the inhibition of Rab7 protein expression by MEHP and had no effect on Sirt1 and Foxo1 protein expression. This suggests that MEHP inhibits lipophagy in TM3 cells by suppressing the Sirt1/Foxo1/Rab7 signaling pathway, ultimately leading to a further decrease in cellular testosterone secretion. This study improves our current understanding of the toxicity and molecular mechanisms of action of MEHP and provides new insights into the reproductive effects of phthalic acid esters.
Collapse
Affiliation(s)
- Yu Hao
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Xuan'en Tian
- Department of Pediatric Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Fengmei Yan
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Xiuqin Wang
- Ningxia Hui Autonomous Region Center for Disease Control and Prevention, Yinchuan, Ningxia 750004, China
| | - Jing Huang
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Ling Li
- Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| |
Collapse
|
10
|
Liu F, Lu Y, Wang X, Sun S, Pan H, Wang M, Wang Z, Zhang W, Ma S, Sun G, Chu Q, Wang S, Qu J, Liu GH. Identification of FOXO1 as a geroprotector in human synovium through single-nucleus transcriptomic profiling. Protein Cell 2024; 15:441-459. [PMID: 38092362 PMCID: PMC11131031 DOI: 10.1093/procel/pwad060] [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: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 05/29/2024] Open
Abstract
The synovium, a thin layer of tissue that is adjacent to the joints and secretes synovial fluid, undergoes changes in aging that contribute to intense shoulder pain and other joint diseases. However, the mechanism underlying human synovial aging remains poorly characterized. Here, we generated a comprehensive transcriptomic profile of synovial cells present in the subacromial synovium from young and aged individuals. By delineating aging-related transcriptomic changes across different cell types and their associated regulatory networks, we identified two subsets of mesenchymal stromal cells (MSCs) in human synovium, which are lining and sublining MSCs, and found that angiogenesis and fibrosis-associated genes were upregulated whereas genes associated with cell adhesion and cartilage development were downregulated in aged MSCs. Moreover, the specific cell-cell communications in aged synovium mirrors that of aging-related inflammation and tissue remodeling, including vascular hyperplasia and tissue fibrosis. In particular, we identified forkhead box O1 (FOXO1) as one of the major regulons for aging differentially expressed genes (DEGs) in synovial MSCs, and validated its downregulation in both lining and sublining MSC populations of the aged synovium. In human FOXO1-depleted MSCs derived from human embryonic stem cells, we recapitulated the senescent phenotype observed in the subacromial synovium of aged donors. These data indicate an important role of FOXO1 in the regulation of human synovial aging. Overall, our study improves our understanding of synovial aging during joint degeneration, thereby informing the development of novel intervention strategies aimed at rejuvenating the aged joint.
Collapse
Affiliation(s)
- Feifei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Lu
- Sports Medicine Department, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Xuebao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuhui Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Huize Pan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Min Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Division of Life Sciences and Medicine, School of Life Sciences, University of Science and Technology of China, Hefei 230001, China
| | - Zehua Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Guoqiang Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qun Chu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- The Fifth People’s Hospital of Chongqing, Chongqing 400062, China
| | - Si Wang
- The Fifth People’s Hospital of Chongqing, Chongqing 400062, China
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
11
|
Xu M, Qian Z, Zhang Y, Gao X, Ma Z, Jin X, Wu S. Sirt1 alleviates osteoarthritis via promoting FoxO1 nucleo-cytoplasm shuttling to facilitate autophagy. Int Immunopharmacol 2024; 131:111893. [PMID: 38513577 DOI: 10.1016/j.intimp.2024.111893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
This study aims to investigate the role and underlying mechanisms of Sirt1 in the pathophysiological process of OA. Safranine O and HE staining were utilized to identify pathological changes in the cartilage tissue. Immunohistochemistry was employed to evaluate the expression levels of proteins. IL-1β treatment and TamCartSirt1flox/flox mice were utilized to induce OA model both in vitro and in vivo. Key autophagy-related transcription factors, autophagy-related genes, and chondrocyte extracellular matrix (ECM) breakdown enzyme markers were examined using multi assays. Immunofluorescence staining revealed subcellular localization and gene expression patterns. ChIP assay and Co-immunoprecipitation assay were conducted to investigate the interactions between FoxO1 and the promoter regions of Atg7 and Sirt1. Our results demonstrate that Sirt1 deficiency exhibited inhibitory effects on ECM synthesis and autophagy, as well as exacerbated angiogenesis. Moreover, Atg7, Foxo1, and Sirt1 could form a protein complex. Sirt1 was observed to facilitate nuclear translocation of FoxO1, enhancing its transcriptional activity. Furthermore, FoxO1 was found to bind to the promoter regions of Atg7 and Sirt1, potentially regulating their expression. This study provides valuable insights into the involvement of Sirt1-Atg7-FoxO1 loop in OA, opening new avenues for targeted therapeutic interventions aiming to mitigate cartilage degradation and restore joint function.
Collapse
Affiliation(s)
- Mao Xu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China; School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhuang Qian
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Ying Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China
| | - Xin Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Zhengmin Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Xinxin Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| | - Shufang Wu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, China.
| |
Collapse
|
12
|
Banjac K, Obradovic M, Zafirovic S, Isenovic ER. Insulin-like growth factor-1 reduces cardiac autosis through decreasing AMPK/FOXO1 signaling and Na +/K +-ATPase-Beclin-1 interaction. Arch Med Sci 2024; 20:1011-1015. [PMID: 39050160 PMCID: PMC11264057 DOI: 10.5114/aoms/177618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/26/2023] [Indexed: 07/27/2024] Open
Abstract
Introduction Insulin-like growth factor-1 (IGF-1) promotes survival and inhibits cardiac autophagy disruption. Methods Male Wistar rats were treated with IGF-1 (50 µg/kg), and 24 h after injection hearts were excised. The level of interaction between Beclin-1 and the α1 subunit of sodium/potassium-adenosine triphosphates (Na+/K+-ATPase), and phosphorylated forms of IGF-1 receptor/insulin receptor (IGF-1R/IR), forkhead box protein O1 (FOXO1) and AMP-activated protein kinase (AMPK) were measured. Results The results indicate that IGF-1 decreased Beclin-1's association with Na+/K+-ATPase (p < 0.05), increased IGF-1R/IR and FOXO1 phosphorylation (p < 0.05), and decreased AMPK phosphorylation (p < 0.01) in rats' hearts. Conclusions The new IGF-1 therapy may control autosis and minimize cardiomyocyte mortality.
Collapse
Affiliation(s)
- Katarina Banjac
- Department of Radiobiology and Molecular Genetics, "VINČA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milan Obradovic
- Department of Radiobiology and Molecular Genetics, "VINČA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sonja Zafirovic
- Department of Radiobiology and Molecular Genetics, "VINČA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, "VINČA" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
13
|
Kim KH, Oprescu SN, Snyder MM, Kim A, Jia Z, Yue F, Kuang S. PRMT5 mediates FoxO1 methylation and subcellular localization to regulate lipophagy in myogenic progenitors. Cell Rep 2023; 42:113329. [PMID: 37883229 PMCID: PMC10727913 DOI: 10.1016/j.celrep.2023.113329] [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: 05/23/2023] [Revised: 08/29/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
Development is regulated by various factors, including protein methylation status. While PRMT5 is well known for its roles in oncogenesis by mediating symmetric di-methylation of arginine, its role in normal development remains elusive. Using Myod1Cre to drive Prmt5 knockout in embryonic myoblasts (Prmt5MKO), we dissected the role of PRMT5 in myogenesis. The Prmt5MKO mice are born normally but exhibit progressive muscle atrophy and premature death. Prmt5MKO inhibits proliferation and promotes premature differentiation of embryonic myoblasts, reducing the number and regenerative function of satellite cells in postnatal mice. Mechanistically, PRMT5 methylates and destabilizes FoxO1. Prmt5MKO increases the total FoxO1 level and promotes its cytoplasmic accumulation, leading to activation of autophagy and depletion of lipid droplets (LDs). Systemic inhibition of autophagy in Prmt5MKO mice restores LDs in myoblasts and moderately improves muscle regeneration. Together, PRMT5 is essential for muscle development and regeneration at least partially through mediating FoxO1 methylation and LD turnover.
Collapse
Affiliation(s)
- Kun Ho Kim
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Stephanie N Oprescu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Madigan M Snyder
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Aran Kim
- Department of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Zhihao Jia
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
14
|
Qin J, Zhang J, Wu JJ, Ru X, Zhong QL, Zhao JM, Lan NH. Identification of autophagy-related genes in osteoarthritis articular cartilage and their roles in immune infiltration. Front Immunol 2023; 14:1263988. [PMID: 38090564 PMCID: PMC10711085 DOI: 10.3389/fimmu.2023.1263988] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Background Autophagy plays a critical role in the progression of osteoarthritis (OA), mainly by regulating inflammatory and immune responses. However, the underlying mechanisms remain unclear. This study aimed to investigate the potential relevance of autophagy-related genes (ARGs) associated with infiltrating immune cells in OA. Methods GSE114007, GSE169077, and ARGs were obtained from the Gene Expression Omnibus (GEO) database and the Human Autophagy database. R software was used to identify the differentially expressed autophagy-related genes (DEARGs) in OA. Functional enrichment and protein-protein interaction (PPI) analyses were performed to explore the role of DEARGs in OA cartilage, and then Cytoscape was utilized to screen hub ARGs. Single-sample gene set enrichment analysis (ssGSEA) was used to conduct immune infiltration analysis and evaluate the potential correlation of key ARGs and immune cell infiltration. Then, the expression levels of hub ARGs in OA were further verified by the GSE169077 and qRT-PCR. Finally, Western blotting and immunohistochemistry were used to validate the final hub ARGs. Results A total of 24 downregulated genes and five upregulated genes were identified, and these genes were enriched in autophagy, mitophagy, and inflammation-related pathways. The intersection results identified nine hub genes, namely, CDKN1A, DDIT3, FOS, VEGFA, RELA, MAP1LC3B, MYC, HSPA5, and HSPA8. GSE169077 and qRT-PCR validation results showed that only four genes, CDKN1A, DDT3, MAP1LC3B, and MYC, were consistent with the bioinformatics analysis results. Western blotting and immunohistochemical (IHC) showed that the expression of these four genes was significantly downregulated in the OA group, which is consistent with the qPCR results. Immune infiltration correlation analysis indicated that DDIT3 was negatively correlated with immature dendritic cells in OA, and FOS was positively correlated with eosinophils. Conclusion CDKN1A, DDIT3, MAP1LC3B, and MYC were identified as ARGs that were closely associated with immune infiltration in OA cartilage. Among them, DDIT3 showed a strong negative correlation with immature dendritic cells. This study found that the interaction between ARGs and immune cell infiltration may play a crucial role in the pathogenesis of OA; however, the specific interaction mechanism needs further research to be clarified. This study provides new insights to further understand the molecular mechanisms of immunity involved in the process of OA by autophagy.
Collapse
Affiliation(s)
- Jun Qin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Clinical Medical Research Center for Orthopedic Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Medical Cosmetology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jin Zhang
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jian-Jun Wu
- Department of Orthopedics, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiao Ru
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Clinical Medical Research Center for Orthopedic Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiu-Ling Zhong
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Clinical Medical Research Center for Orthopedic Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jin-Min Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Clinical Medical Research Center for Orthopedic Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Research Centre for Regenerative Medicine, Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ni-Han Lan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Clinical Medical Research Center for Orthopedic Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
15
|
Yu F, Wang M, Luo K, Sun L, Yu S, Zuo J, Wang Y. Expression Profiles of Long Non-Coding RNAs in the Articular Cartilage of Rats Exposed to T-2 Toxin. Int J Mol Sci 2023; 24:13703. [PMID: 37762015 PMCID: PMC10530968 DOI: 10.3390/ijms241813703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
T-2 toxin could induce bone damage. But there is no specific mechanism about the long non-coding RNAs (lncRNAs) involved in T-2 toxin-induced articular cartilage injury. In this study, 24 SD rats were randomly divided into a control group and a T-2 group, which were administered 4% absolute ethanol and 100 ng/g · bw/day of T-2 toxin, respectively. After treatment for 4 weeks, safranin O/fast green staining identified the pathological changes in the articular cartilage of rats, and immunofluorescence verified the autophagy level increase in the T-2 group. Total RNA was isolated, and high-throughput sequencing was performed. A total of 620 differentially expressed lncRNAs (DE-lncRNAs) were identified, and 326 target genes were predicted. Enrichment analyses showed that the target genes of DE-lncRNAs were enriched in the autophagy-related biological processes and pathways. According to the autophagy database, a total of 23 autophagy-related genes were identified, and five hub genes (Foxo3, Foxo1, Stk11, Hdac4, and Rela) were screened using the Maximal Clique Centrality algorithm. The Human Protein Atlas database indicated that Rela and Hdac4 proteins were highly expressed in the bone marrow tissue, while Foxo3, Foxo1, and Stk11 proteins were reduced. According to Enrichr, etoposide and diatrizoic acid were identified as the key drugs. The real-time quantitative PCR results were consistent with the RNA sequencing (RNA-Seq) results. These results suggested that autophagy was involved in the rat articular cartilage lesions induced by T-2 toxin. The lncRNAs of NONRATG014223.2, NONRATG012484.2, NONRATG021591.2, NONRATG024691.2, and NONRATG002808.2, and their target genes of Foxo3, Foxo1, Stk11, Hdac4, and Rela, respectively, were the key regulator factors of autophagy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yanjie Wang
- School of Public Health, Zhengzhou University, Zhengzhou 450001, China; (F.Y.); (M.W.); (K.L.); (L.S.); (S.Y.); (J.Z.)
| |
Collapse
|
16
|
Wu W, He X, Li S, Jin M, Ni Y. Pain nursing for gynecologic cancer patients. Front Oncol 2023; 13:1205553. [PMID: 37564934 PMCID: PMC10410261 DOI: 10.3389/fonc.2023.1205553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Gynecological malignancy remains a prevalent cause of mortality among women. Chronic cancer pain, as a severe complication of malignancy and its therapies, accounts for a substantial burden of physical and psychological distress in affected patients. Accordingly, early identification, assessment, and standardized management of such pain are crucial in the prevention or delay of its progression. In the present review, we provide a comprehensive overview of the pathological factors that contribute to pain in patients with gynecological malignancy while highlighting the underlying mechanisms of pain in this population. In addition, we summarize several treatment modalities targeting pain management in gynecologic cancer patients, including surgery, radiotherapy, and chemotherapy. These interventions are crucial for tumor elimination and patient survival. Chronic cancer pain exerts a significant impact on wellbeing and quality of life for patients with gynecologic cancer. Therefore, our review emphasizes the importance of addressing this pain and its psychological sequelae and advocates for a multidisciplinary approach that encompasses nursing and psychological support. In summary, this review offers valuable insights into the pathological factors underlying pain, reviews pain management modalities, and stresses the critical role of early intervention and comprehensive care in enhancing the quality of life of these patients.
Collapse
Affiliation(s)
| | - Xiaodan He
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | | | | | | |
Collapse
|
17
|
Liu Y, Zhang Z, Liu C, Zhang H. Sirtuins in osteoarthritis: current understanding. Front Immunol 2023; 14:1140653. [PMID: 37138887 PMCID: PMC10150098 DOI: 10.3389/fimmu.2023.1140653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Osteoarthritis (OA) is a common disease characterized by severe chronic joint pain, that imposes a large burden on elderly people. OA is a highly heterogeneous disease, and multiple etiologies contribute to its progression. Sirtuins (SIRTs) are Class III histone deacetylases (HDACs) that regulate a comprehensive range of biological processes such as gene expression, cell differentiation, and organism development, and lifespan. Over the past three decades, increasing evidence has revealed that SIRTs are not only important energy sensors but also protectors against metabolic stresses and aging, and an increasing number of studies have focused on the functions of SIRTs in OA pathogenesis. In this review, we illustrate the biological functions of SIRTs in OA pathogenesis from the perspectives of energy metabolism, inflammation, autophagy and cellular senescence. Moreover, we offer insights into the role played by SIRTs in regulating circadian rhythm, which has recently been recognized to be crucial in OA development. Here, we provide the current understanding of SIRTs in OA to guide a new direction for OA treatment exploration.
Collapse
|