1
|
Shi J, Chen L, Wang X, Ma X. KLF2 Inhibits Ferroptosis and Improves Mitochondrial Dysfunction in Chondrocyte Through SIRT1/GPX4 Signaling to Improve Osteoarthritis. Drug Dev Res 2024; 85:e70015. [PMID: 39527654 DOI: 10.1002/ddr.70015] [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: 09/03/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Osteoarthritis (OA), a disease of articular joints, is the leading cause of disability in the elderly. Repressing ferroptosis and improving mitochondrial function can delay the progression of OA. Kruppel-like factor 2 (KLF2) exerts a protective effect on OA. However, whether KLF2 affects ferroptosis and mitochondrial function during OA remains unknown. The OA in vivo and in vitro models were constructed in this work. The structural damage of knee joint in OA mice was evaluated through Micro-CT scanning. H&E, SOFG, TB, and TUNEL staining were applied for pathological examination of cartilage tissues. ELISA was employed to examine the contents of inflammatory factors. Additionally, iron deposition in cartilage tissues was measured by Prussian blue staining, and the levels of proteins related to ferroptosis were assessed by immunoblotting. Besides, mitochondrial morphology and function were estimated using a transmission electron microscope and JC-1 staining. In interleukin (IL)-1β-treated C28/I2 cells, the levels of inflammatory factors, intracellular ROS, mitochondrial ROS, lipid ROS, and Fe2+ were measured. Mitochondrial function was evaluated by detecting the levels of mitochondrial membrane potential (MMP), ATP, mPTP, and OCR. KLF2 overexpression ameliorated the structural damage of knee cartilage in OA mice. KLF2 upregulation inhibited ferroptosis and alleviated mitochondrial damage in knee cartilage of OA mice and IL-1β-treated C28/I2 cells. Moreover, KLF2 overexpression activated SIRT1/GPX4 signaling in vivo and in vitro. EX527 addition blocked the influences of KLF2 upregulation on ferroptosis and mitochondrial dysfunction in IL-1β-treated C28/I2 cells. Altogether KLF2 inhibits ferroptosis and improves mitochondrial dysfunction in chondrocytes through SIRT1/GPX4 signaling to improve OA.
Collapse
Affiliation(s)
- Jiaqi Shi
- Orthopedic Department, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Li Chen
- Orthopedic Department, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xu Wang
- Orthopedic Department, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xin Ma
- Orthopedic Department, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
2
|
Liu XY, Yu QP, Guo SQ, Chen XM, Zeng WN, Zhou ZK. High expression of transcription factor EGR1 is associated with postoperative muscle atrophy in patients with knee osteoarthritis undergoing total knee arthroplasty. J Orthop Surg Res 2024; 19:618. [PMID: 39354574 PMCID: PMC11443708 DOI: 10.1186/s13018-024-05109-9] [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: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Muscle atrophy is a typical affliction in patients affected by knee Osteoarthritis (KOA). This study aimed to examine the potential pathogenesis and biomarkers that coalesce to induce muscle atrophy, primarily through the utilization of bioinformatics analysis. METHODS Two distinct public datasets of osteoarthritis and muscle atrophy (GSE82107 and GSE205431) were subjected to differential gene expression analysis and gene set enrichment analysis (GSEA) to probe for common differentially expressed genes (DEGs) and conduct transcription factor (TF) enrichment analysis from such genes. Venn diagrams were used to identify the target TF, followed by the construction of a protein-protein interaction (PPI) network of the common DEGs governed by the target TF. Hub genes were determined through the CytoHubba plug-in whilst their biological functions were assessed using GSEA analysis in the GTEx database. To validate the study, reverse transcriptase real-time quantitative polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and Flow Cytometry techniques were employed. RESULTS A total of 138 common DEGs of osteoarthritis and muscle atrophy were identified, with 16 TFs exhibiting notable expression patterns in both datasets. Venn diagram analysis identified early growth response gene-1 (EGR1) as the target TF, enriched in critical pathways such as epithelial mesenchymal transition, tumor necrosis factor-alpha signaling NF-κB, and inflammatory response. PPI analysis revealed five hub genes, including EGR1, FOS, FOSB, KLF2, and JUNB. The reliability of EGR1 was confirmed by validation testing, corroborating bioinformatics analysis trends. CONCLUSIONS EGR1, FOS, FOSB, KLF2, and JUNB are intricately involved in muscle atrophy development. High EGR1 expression directly regulated these hub genes, significantly influencing postoperative muscle atrophy progression in KOA patients.
Collapse
Grants
- 2023HXFH012 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University
- 2023HXFH012 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University
- 2023HXFH012 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University
- 2023HXFH012 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University
- 2023HXFH012 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University
- 2023HXFH012 1·3·5 project for disciplines of excellence, West China Hospital, Sichuan University
Collapse
Affiliation(s)
- Xiao-Yang Liu
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Qiu-Ping Yu
- Health Management Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Si-Qin Guo
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Xu-Ming Chen
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Wei-Nan Zeng
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Zong-Ke Zhou
- Department of Orthopedics, Orthopaedic Research Institute, West China Hospital, Sichuan University, #37 Guoxue Road, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
3
|
Xiang T, Yang C, Deng Z, Sun D, Luo F, Chen Y. Krüppel-like factors family in health and disease. MedComm (Beijing) 2024; 5:e723. [PMID: 39263604 PMCID: PMC11387732 DOI: 10.1002/mco2.723] [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: 03/19/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024] Open
Abstract
Krüppel-like factors (KLFs) are a family of basic transcription factors with three conserved Cys2/His2 zinc finger domains located in their C-terminal regions. It is acknowledged that KLFs exert complicated effects on cell proliferation, differentiation, survival, and responses to stimuli. Dysregulation of KLFs is associated with a range of diseases including cardiovascular disorders, metabolic diseases, autoimmune conditions, cancer, and neurodegenerative diseases. Their multidimensional roles in modulating critical pathways underscore the significance in both physiological and pathological contexts. Recent research also emphasizes their crucial involvement and complex interplay in the skeletal system. Despite the substantial progress in understanding KLFs and their roles in various cellular processes, several research gaps remain. Here, we elucidated the multifaceted capabilities of KLFs on body health and diseases via various compliable signaling pathways. The associations between KLFs and cellular energy metabolism and epigenetic modification during bone reconstruction have also been summarized. This review helps us better understand the coupling effects and their pivotal functions in multiple systems and detailed mechanisms of bone remodeling and develop potential therapeutic strategies for the clinical treatment of pathological diseases by targeting the KLF family.
Collapse
Affiliation(s)
- Tingwen Xiang
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Chuan Yang
- Department of Biomedical Materials Science Third Military Medical University (Army Medical University) Chongqing China
| | - Zihan Deng
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Dong Sun
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Fei Luo
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
| | - Yueqi Chen
- Department of Orthopedics Southwest Hospital Third Military Medical University (Army Medical University) Chongqing China
- Department of Orthopedics Chinese PLA 76th Army Corps Hospital Xining China
| |
Collapse
|
4
|
Grol MW. The evolving landscape of gene therapy strategies for the treatment of osteoarthritis. Osteoarthritis Cartilage 2024; 32:372-384. [PMID: 38199296 DOI: 10.1016/j.joca.2023.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/05/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
OBJECTIVES Significant advances have been made in our understanding of osteoarthritis (OA) pathogenesis; however, no disease-modifying therapies have been identified. This review will summarize the gene therapy landscape, its initial successes for OA, and possible challenges using recent studies and examples of gene therapies in clinical trials. DESIGN This narrative review has three major sections: 1) vector systems for OA gene therapy, 2) current and emerging targets for OA gene therapy, and 3) considerations and future directions. RESULTS Gene therapy is the strategy by which nucleic acids are delivered to treat and reverse disease progression. Specificity and prolonged expression of these nucleic acids are achieved by manipulating promoters, genes, and vector systems. Certain vector systems also allow for the development of combinatorial nucleic acid strategies that can be delivered in a single intraarticular injection - an approach likely required to treat the complexity of OA pathogenesis. Several viral and non-viral vector-based gene therapies are in clinical trials for OA, and many more are being evaluated in the preclinical arena. CONCLUSIONS In a post-coronavirus disease 2019 (COVID-19) era, the future of gene therapy for OA is certainly promising; however, the majority of preclinical validation continues to focus heavily on post-traumatic models and changes in only cartilage and subchondral bone. To ensure successful translation, new candidates in the preclinical arena should be examined against all joint tissues as well as pain using diverse models of injury-, obesity-, and age-induced disease. Lastly, consideration must be given to strategies for repeat administration and the cost of treatment owing to the chronic nature of OA.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
5
|
Yang X, Dong S, Fan Y, Xia Y, Yang F, Chen Z, Chen D, Zhang M, Liang D, Zeng C. Krüppel-like Factor 15 Suppresses Ferroptosis by Activating an NRF2/GPX4 Signal to Protect against Folic Acid-Induced Acute Kidney Injury. Int J Mol Sci 2023; 24:14530. [PMID: 37833977 PMCID: PMC10572468 DOI: 10.3390/ijms241914530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/16/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Acute kidney injury (AKI) is a common and serious disease with high morbidity and mortality, and its pathophysiological mechanisms are not fully understood. Increasing evidence suggests an important role of ferroptosis in AKI. Krüppel-like factor 15 (KLF15) is a transcription factor involved in several metabolic diseases, but its role in AKI and ferroptosis remains unclear. In this study, we explored the potential role of KLF15 using a folic acid-induced AKI model. Our study showed that KLF15 expression was reduced in kidney tissues of AKI mice, and KLF15 knockout exacerbated folic acid-induced ferroptosis and kidney injury. In vitro studies revealed that the ferroptosis inducer erastin significantly suppressed KLF15 expression in human tubular epithelial cells. Notably, the overexpression of KLF15 attenuated ferroptosis, as evidenced by a decrease in the lipid peroxidation marker of malondialdehyde and the upregulation of glutathione peroxidase 4 (GPX4), while KLF15 knockdown with shRNA exerted the opposite effect. Mechanistically, KLF15 stabilized the protein of nuclear factor erythroid 2-related factor 2 (NRF2) and subsequently increased the GPX4 level. Collectively, KLF15 plays an important role in the modulation of ferroptosis in AKI and may be a potential therapeutic target for treating AKI.
Collapse
Affiliation(s)
- Xue Yang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Shihui Dong
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Yun Fan
- Jinling Clinical Medical College, Nanjing Medical University, Nanjing 210008, China
| | - Yuanyuan Xia
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Fan Yang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Zhaohong Chen
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Dacheng Chen
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Mingchao Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Dandan Liang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| | - Caihong Zeng
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210044, China
| |
Collapse
|
6
|
Chen F, Zhan J, Liu M, Mamun AA, Huang S, Tao Y, Zhao J, Zhang Y, Xu Y, He Z, Du S, Lu W, Li X, Chen Z, Xiao J. FGF2 Alleviates Microvascular Ischemia-Reperfusion Injury by KLF2-mediated Ferroptosis Inhibition and Antioxidant Responses. Int J Biol Sci 2023; 19:4340-4359. [PMID: 37705747 PMCID: PMC10496511 DOI: 10.7150/ijbs.85692] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/08/2023] [Indexed: 09/15/2023] Open
Abstract
An essential pathogenic element of acute limb ischemia/reperfusion (I/R) injury is microvascular dysfunction. The majority of studies indicates that fibroblast growth factor 2 (FGF2) exhibits protective properties in cases of acute I/R injury. Albeit its specific role in the context of acute limb I/R injury is yet unknown. An impressive post-reperfusion increase in FGF2 expression was seen in a mouse model of hind limb I/R, followed by a decline to baseline levels, suggesting a key role for FGF2 in limb survivability. FGF2 appeared to reduce I/R-induced hypoperfusion, tissue edema, skeletal muscle fiber injury, as well as microvascular endothelial cells (ECs) damage within the limb, according to assessments of limb vitality, Western blotting, and immunofluorescence results. The bioinformatics analysis of RNA-sequencing revealed that ferroptosis played a key role in FGF2-facilitated limb preservation. Pharmacological inhibition of NFE2L2 prevented ECs from being affected by FGF2's anti-oxidative and anti-ferroptosis activities. Additionally, silencing of kruppel-like factor 2 (KLF2) by interfering RNA eliminated the antioxidant and anti-ferroptosis effects of FGF2 on ECs. Further research revealed that the AMPK-HDAC5 signal pathway is the mechanism via which FGF2 regulates KLF2 activity. Data from luciferase assays demonstrated that overexpression of HDAC5 prevented KLF2 from becoming activated by FGF2. Collectively, FGF2 protects microvascular ECs from I/R injury by KLF2-mediated ferroptosis inhibition and antioxidant responses.
Collapse
Affiliation(s)
- Fanfeng Chen
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Jiayu Zhan
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Mi Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Shanshan Huang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yibing Tao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Jiaxin Zhao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Yitie Xu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Zili He
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Shenghu Du
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Wei Lu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xiaokun Li
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| | - Zimiao Chen
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Jian Xiao
- Department of Wound healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
7
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
8
|
Liu W, Zhang F, Liang W, Huang K, Jia C, Zhang J, Li X, Wei W, Gong R, Chen J. Integrated insight into the molecular mechanisms of selenium-modulated, MPP +-induced cytotoxicity in a Parkinson's disease model. J Trace Elem Med Biol 2023; 79:127208. [PMID: 37269647 DOI: 10.1016/j.jtemb.2023.127208] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/13/2022] [Accepted: 05/17/2023] [Indexed: 06/05/2023]
Abstract
OBJECTIVE Parkinson's disease (PD) is a neurodegenerative disease that is associated with oxidative stress. Due to the anti-inflammatory and antioxidant functions of Selenium (Se), this molecule may have neuroprotective functions in PD; however, the involvement of Se in such a protective function is unclear. METHODS 1-methyl-4-phenylpyridinium (MPP+), which inhibits mitochondrial respiration, is generally used to produce a reliable cellular model of PD. In this study, a MPP+-induced PD model was used to test if Se could modulate cytotoxicity, and we further capture gene expression profiles following PC12 cell treatment with MPP+ with or without Se by genome wide high-throughput sequencing. RESULTS We identified 351 differentially expressed genes (DEGs) and 14 differentially expressed long non-coding RNAs (DELs) in MPP+-treated cells when compared to controls. We further document 244 DEGs and 27 DELs in cells treated with MPP+ and Se vs. cells treated with MPP+ only. Functional annotation analysis of DEGs and DELs revealed that these groups were enriched in genes that respond to reactive oxygen species (ROS), metabolic processes, and mitochondrial control of apoptosis. Thioredoxin reductase 1 (Txnrd1) was also identified as a biomarker of Se treatment. CONCLUSIONS Our data suggests that the DEGs Txnrd1, Siglec1 and Klf2, and the DEL AABR07044454.1 which we hypothesize to function in cis on the target gene Cdkn1a, may modulate the underlying neurodegenerative process, and act a protective function in the PC12 cell PD model. This study further systematically demonstrated that mRNAs and lncRNAs induced by Se are involved in neuroprotection in PD, and provides novel insight into how Se modulates cytotoxicity in the MPP+-induced PD model.
Collapse
Affiliation(s)
- Wen Liu
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China
| | - Feiyang Zhang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China
| | - Wu Liang
- Department of Neurosurgery, Minda Hospital of Hubei Minzu University, Enshi 445000, China
| | - Kaixin Huang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China
| | - Chenguang Jia
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China
| | - Jie Zhang
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China; Neuroepigenetic Research Lab, Medical Research Institute, Wuhan University, Donghu Road 115, Wuhan 430071, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China; Neuroepigenetic Research Lab, Medical Research Institute, Wuhan University, Donghu Road 115, Wuhan 430071, China.
| | - Rui Gong
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China.
| | - Jincao Chen
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Donghu Road 169, Wuhan 430071, China.
| |
Collapse
|
9
|
Hu H, Yang J, Zhong Y, Wang J, Cai J, Luo C, Jin Z, Gao M, He M, Zheng L. Polydopamine-Pd nanozymes as potent ROS scavengers in combination with near-infrared irradiation for osteoarthritis treatment. iScience 2023; 26:106605. [PMID: 37182095 PMCID: PMC10172781 DOI: 10.1016/j.isci.2023.106605] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/27/2022] [Accepted: 03/29/2023] [Indexed: 05/16/2023] Open
Abstract
Excessive reactive oxygen species (ROS) in joints could lead to gradual degeneration of the extracellular matrix (ECM) and apoptosis of chondrocytes, contributing to the occurrence and development of osteoarthritis (OA). Mimicking natural enzymes, polydopamine (PDA)-based nanozymes showed great potential in treating various inflammatory diseases. In this work, PDA loaded with ultra-small palladium (PDA-Pd) nanoparticles (NPs) was employed to scavenge ROS for OA therapy. As a result, PDA-Pd effectively declined the intracellular ROS levels and exhibited efficient antioxidative and anti-inflammatory capacity with good biocompatibility in IL-1β stimulated chondrocytes. Significantly, assisted with near-infrared (NIR) irradiation, its therapeutic effect was further enhanced. Further, NIR-stimulated PDA-Pd suppressed the progression of OA after intra-articular injection in the OA rat model. With favorable biocompatibility, PDA-Pd exhibits efficient antioxidative and anti-inflammatory capacity, leading to the alleviation of OA in rats. Our findings may provide new insights into the treatment of various ROS-induced inflammatory diseases.
Collapse
Affiliation(s)
- Hao Hu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Junxu Yang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yanping Zhong
- Life Sciences Institute of Guangxi Medical University, Nanning 530021, China
| | - Jiawei Wang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinhong Cai
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Cuijuan Luo
- Life Sciences Institute of Guangxi Medical University, Nanning 530021, China
| | - Zhiqiang Jin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ming Gao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Corresponding author
| | - Maolin He
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Corresponding author
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by the Province and Ministry, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Corresponding author
| |
Collapse
|
10
|
Tusa I, Menconi A, Tubita A, Rovida E. Pathophysiological Impact of the MEK5/ERK5 Pathway in Oxidative Stress. Cells 2023; 12:cells12081154. [PMID: 37190064 DOI: 10.3390/cells12081154] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Oxidative stress regulates many physiological and pathological processes. Indeed, a low increase in the basal level of reactive oxygen species (ROS) is essential for various cellular functions, including signal transduction, gene expression, cell survival or death, as well as antioxidant capacity. However, if the amount of generated ROS overcomes the antioxidant capacity, excessive ROS results in cellular dysfunctions as a consequence of damage to cellular components, including DNA, lipids and proteins, and may eventually lead to cell death or carcinogenesis. Both in vitro and in vivo investigations have shown that activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway is frequently involved in oxidative stress-elicited effects. In particular, accumulating evidence identified a prominent role of this pathway in the anti-oxidative response. In this respect, activation of krüppel-like factor 2/4 and nuclear factor erythroid 2-related factor 2 emerged among the most frequent events in ERK5-mediated response to oxidative stress. This review summarizes what is known about the role of the MEK5/ERK5 pathway in the response to oxidative stress in pathophysiological contexts within the cardiovascular, respiratory, lymphohematopoietic, urinary and central nervous systems. The possible beneficial or detrimental effects exerted by the MEK5/ERK5 pathway in the above systems are also discussed.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
11
|
Shen J, Lin X, Lin Y, Xiao J, Wu C, Zheng F, Wu X, Lin H, Chen G, Liu H. Supplementation of hyaluronic acid injections with vitamin D improve knee function by attenuating synovial fluid oxidative stress in osteoarthritis patients with vitamin D insufficiency. Front Nutr 2023; 10:1026722. [PMID: 37081922 PMCID: PMC10112517 DOI: 10.3389/fnut.2023.1026722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
ObjectivesThere is still controversy about the effect of vitamin D supplementation on osteoarthritis (OA). The purpose of this study was to investigate the effects of vitamin D supplementation with Hyaluronic acid (HA) injection on OA.MethodsWe investigated serum vitamin D levels and oxidative stress (OS) in synovial fluid from patients with OA who underwent total knee arthroplasty (grade IV, n = 24) and HA injection (grade II and III, n = 40). The effects of HA injection with or without oral vitamin D supplementation on synovial fluid OS and knee pain and function were then further investigated. Finally, patients underwent HA injection were divided into two groups according to vitamin D levels (vitamin D < or > 30 ng/ml), and the efficacy of the two groups were compared.ResultsThe results showed that the levels of glutathione peroxidase (GSH-PX) (P < 0.05) in the synovial fluid were lower in patients with stage IV OA than that in patients with stage II-III OA, while the levels of malondialdehyde (MDA) (P < 0.05) and lactate dehydrogenase (LDH) (P < 0.01) were significantly higher. Moreover, we found that age, BMI and vitamin D levels were significantly associated with the levels of oxidants and/or antioxidants in synovial fluid, and that vitamin D was significantly negatively correlated with BMI (R = −0.3527, p = 0.0043). Supplementation of HA injections with vitamin D significantly reduced the OS status in synovial fluid, attenuated knee pain and improved knee function in OA patients with vitamin D insufficiency.ConclusionWe conclude that maintenance of vitamin D sufficiency may be beneficial for the treatment of OA by improving OS in synovial fluid.
Collapse
|
12
|
Wang L, Liu X. An oxidative stress-related signature for predicting the prognosis of liver cancer. Front Genet 2023; 13:975211. [PMID: 36685933 PMCID: PMC9845401 DOI: 10.3389/fgene.2022.975211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: This study aimed to screen for oxidative stress-related genes (OSRGs) and build an oxidative stress-related signature to predict the prognosis of liver cancer. Methods: OSRGs with a protein domain correlation score ≥ 6 were downloaded from the GeneCards database and intersected with The Cancer Genome Atlas (TCGA) data for subsequent analyses. Differential immune cells (DICs) and immune and stromal scores between the normal and tumor samples were determined, followed by unsupervised hierarchical cluster analysis. Immune-related OSRGs were identified using weighted gene co-expression network analysis. An OSRG-related risk signature was then built, and the GSE14520 dataset was used for validation. A nomogram evaluation model was used to predict prognosis. Results: Nine DICs were determined between the normal and tumor groups, and three subtypes were obtained: clusters 1, 2, and 3. Cluster 1 had the best prognosis among the clusters. One hundred thirty-eight immune-related OSRGs were identified, and seven prognosis-related OSRGs were used to build the OSRG score prognostic model. Patients in the high OSRG score group had a poorer prognosis than those in the low OSRG score group. Six immune cell infiltration and enrichment scores of the 16 immune gene sets showed significant differences between the high and low OSRG score groups. Moreover, a nomogram was constructed based on the prognostic signature and clinicopathological features and had a robust predictive performance and high accuracy. Conclusion: The OSRG-related risk signature and the prognostic nomogram accurately predicted patient survival.
Collapse
|
13
|
Liang J, Wang S, Hu J, Hong X, Zhu M, Liu X, Alswadeh M, Mo F, Dai M. Targeted inhibition of TXNRD1 prevents cartilage extracellular matrix degeneration by activating Nrf2 pathway in osteoarthritis. Biochem Biophys Res Commun 2022; 635:267-276. [PMID: 36308906 DOI: 10.1016/j.bbrc.2022.10.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis, a prevalent orthopedic disease, can affect the elderly and causes impairment. The degradation and aberrant homeostasis of cartilage extracellular matrix figure pivotally in the progression of osteoarthritis. Thioredoxin systems plays a role in a wide range of biological processes, including cell proliferation, apoptosis, and oxidative stress. The present study aimed to investigate the unique function and underlying pathophysiological mechanism of TXNRD1 in chondrocytes. An upregulated expression of TXNRD1 was observed in the articular cartilage of osteoarthritis patients compared with normal articular cartilage. Furthermore, in vitro experiments showed that the expression of TXNRD1 was also abnormally increased in IL-1β-induced primary mouse chondrocytes. Silencing TXNRD1 using siRNA in chondrocytes could effectively inhibit the expression of ADAMTS5 and MMP13, and enhance the expression of COL2A1 and SOX9. The same was true for auranofin, an inhibitor of TXNRD1. This phenomenon indicated that inhibition of TXNRD1 attenuated il-1β-induced metabolic imbalance of extracellular matrix (ECM) and the progression of chondrocyte osteoarthritis. Further mechanism analysis revealed that the activation of Nrf2 signaling pathway and the expression of heme oxygenase-1 (HO-1) were increased upon TXNRD1 inhibition. Furthermore, auranofin was found to attenuate DMM-induced osteoarthritis progression in vivo. Therefore, the pharmacological downregulation of TXNRD1 may provide an effective novel therapy for OA.
Collapse
Affiliation(s)
- Jianhui Liang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Song Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Jiawei Hu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xin Hong
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Meisong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xuqiang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Momen Alswadeh
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Fengbo Mo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China.
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China.
| |
Collapse
|
14
|
Kruppel-like Factors in Skeletal Physiology and Pathologies. Int J Mol Sci 2022; 23:ijms232315174. [PMID: 36499521 PMCID: PMC9741390 DOI: 10.3390/ijms232315174] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/11/2022] Open
Abstract
Kruppel-like factors (KLFs) belong to a large group of zinc finger-containing transcription factors with amino acid sequences resembling the Drosophila gap gene Krüppel. Since the first report of molecular cloning of the KLF family gene, the number of KLFs has increased rapidly. Currently, 17 murine and human KLFs are known to play crucial roles in the regulation of transcription, cell proliferation, cellular differentiation, stem cell maintenance, and tissue and organ pathogenesis. Recent evidence has shown that many KLF family molecules affect skeletal cells and regulate their differentiation and function. This review summarizes the current understanding of the unique roles of each KLF in skeletal cells during normal development and skeletal pathologies.
Collapse
|
15
|
Jiang Z, Wang H, Zhang Z, Pan J, Yuan H. Cartilage targeting therapy with reactive oxygen species-responsive nanocarrier for osteoarthritis. J Nanobiotechnology 2022; 20:419. [PMID: 36123746 PMCID: PMC9484188 DOI: 10.1186/s12951-022-01629-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/10/2022] [Indexed: 11/12/2022] Open
Abstract
Targeting cartilage is a promising strategy for the treatment of osteoarthritis, and various delivery vehicles were developed to assist the therapeutic agents into cartilage. However, the underlying biomechanisms and potential bioactivities remain oversimplified. Inspired by oxidative stress in the pathogenesis of osteoarthritis, we firstly testified the antioxidant capacity of a synthetic small molecule compound, oltipraz (OL), to the chondrocytes treated by IL-1β. Then a functional reactive oxygen species (ROS) responsive nanocarrier, mesoporous silica nanoparticles (MSN) modified with methoxy polyethylene glycol-thioketal, was constructed. In vitro biomolecular results showed that compared with OL alone, MSN-OL could significantly activate Nrf2/HO-1 signaling pathway, which exhibited better ROS-scavenging proficiency and greater anti-apoptotic ability to protect mitochondrial membrane potential of chondrocytes. Further bioinformatics analysis revealed that MSN-OL suppressed clusters of genes associated with extracellular matrix organization, cell apoptosis and cellular response to oxidative stress. Animal experiments further confirmed the great cartilage-protecting ability of MSN-OL through upregulating the expression of Nrf2/HO-1 signaling pathway without obvious toxicity. In summary, this study provided a delivery system through ROS-responsive regulation of the therapeutic agents into chondrocytes of the cartilage, and confirmed the exact biological mechanisms of this innovative strategy.
Collapse
Affiliation(s)
- Zengxin Jiang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hao Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zeng Zhang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jianfeng Pan
- Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Hengfeng Yuan
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
16
|
Song Y, Wu Z, Zhao P. The effects of metformin in the treatment of osteoarthritis: Current perspectives. Front Pharmacol 2022; 13:952560. [PMID: 36081941 PMCID: PMC9445495 DOI: 10.3389/fphar.2022.952560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
Osteoarthritis is a chronic and irreversible disease of the locomotor system which is closely associated with advancing age. Pain and limited mobility frequently affect the quality of life in middle-aged and older adults. With a global population of more than 350 million, osteoarthritis is becoming a health threat alongside cancer and cardiovascular disease. It is challenging to find effective treatments to promote cartilage repair and slow down disease progression. Metformin is the first-line drug for patients with type 2 diabetes, and current perspectives suggest that it cannot only lower glucose but also has anti-inflammatory and anti-aging properties. Experimental studies applying metformin for the treatment of osteoarthritis have received much attention in recent years. In our review, we first presented the history of metformin and the current status of osteoarthritis, followed by a brief review of the mechanism that metformin acts, involving AMPK-dependent and non-dependent pathways. Moreover, we concluded that metformin may be beneficial in the treatment of osteoarthritis by inhibiting inflammation, modulating autophagy, antagonizing oxidative stress, and reducing pain levels. Finally, we analyzed the relevant evidence from animal and human studies. The potential of metformin for the treatment of osteoarthritis deserves to be further explored.
Collapse
|
17
|
Li B, Yang B, Liu X, Zhao J, Ross RP, Stanton C, Zhang H, Chen W. Microbiota-assisted therapy for systemic inflammatory arthritis: advances and mechanistic insights. Cell Mol Life Sci 2022; 79:470. [PMID: 35932328 PMCID: PMC11072763 DOI: 10.1007/s00018-022-04498-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/22/2022]
Abstract
Research on the influence of gut microbiota on systemic inflammatory arthritis has exploded in the past decade. Gut microbiota changes may be a crucial regulatory component in systemic inflammatory arthritis. As a result of advancements in the field, microbiota-assisted therapy has evolved, but this discipline is still in its infancy. Consequently, we review the limitations of current systemic inflammatory arthritis treatment, analyze the connection between the microbiota and arthritis, and summarize the research progress of microbiota regulating systemic inflammatory arthritis and the further development aspects of microbiota-assisted therapy. Finally, the partial mechanisms of microbiota-assisted therapy of systemic inflammatory arthritis are being discussed. In general, this review summarizes the current progress, challenges, and prospects of microbiota-assisted therapy for systemic inflammatory arthritis and points out the direction for the development of microbiota-assisted therapy in the future.
Collapse
Affiliation(s)
- Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - R Paul Ross
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Catherine Stanton
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China.
- Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China.
| |
Collapse
|
18
|
CDDO-Im ameliorates osteoarthritis and inhibits chondrocyte apoptosis in mice via enhancing Nrf2-dependent autophagy. Acta Pharmacol Sin 2022; 43:1793-1802. [PMID: 34754093 PMCID: PMC9253092 DOI: 10.1038/s41401-021-00782-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic degenerative joint disease with few treatment options. The pathogenesis of OA is characterized by sustained inflammation, oxidative stress and chondrocyte apoptosis that eventually lead to cartilage degradation and joint dysfunction. In the present study, we identified a synthetic triterpenoid CDDO-Im(1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl] imidazole) as an activator of Nrf2 (nuclear factor erythroid 2-related factor 2) that displayed strong anti-OA effects. We showed that CDDO-Im (20 nM) significantly alleviated TNF-α-induced apoptosis of primary human chondrocytes and extracellular matrix degradation. In a mouse OA model incurred by DMM (destabilization of medial meniscus), administration of CDDO-Im (2.5 mg/kg, ip, every other day for 8 weeks) effectively reduced knee joint cartilage erosion and serum levels of inflammatory cytokines IL-1β and IL-6. We revealed that CDDO-Im (20 nM) significantly enhanced autophagy activities in chondrocytes, whereas the autophagy inhibition by chloroquine (CQ, 50 μM) or 3-methyladenine (3-MA, 5 mM) abrogated the anti-apoptosis and chondroprotective effects of CDDO-Im in TNF-α-treated chondrocytes. Moreover, we confirmed that CDDO-Im (1-20 nM) dose-dependently activated Nrf2 pathway in TNF-α-treated chondrocytes, and its chondroprotective and autophagy-enhancing effects were significantly diminished when Nrf2 signaling was blocked by Nrf2 inhibitor ML385 (20 μM) or siRNA-mediated Nrf2 knockdown. Together, our results demonstrate that CDDO-Im exhibits prominent chondroprotective and anti-OA activities owing to its Nrf2 activation and autophagy-enhancing properties, which might provide new insights into the strategies of OA clinical prevention and treatment.
Collapse
|
19
|
Protective Effects of Cannabidiol on Chemotherapy-Induced Oral Mucositis via the Nrf2/Keap1/ARE Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4619760. [PMID: 35669853 PMCID: PMC9165619 DOI: 10.1155/2022/4619760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Oral mucositis (OM) is a common complication during chemotherapy characterized by ulceration, mucosa atrophy, and necrosis, which seriously interferes with nutritional intake and oncotherapy procedures among patients. However, the efficacy of current treatments for OM remains limited. Cannabidiol (CBD) is a natural cannabinoid with multiple biological activities, including antioxidant and anti-inflammatory potential. In this study, we aimed to investigate the chemopreventive effects and mechanisms of CBD in protecting C57BL/6N mice and human oral keratinocytes (HOK) from 5-fluorouracil- (5-FU-) induced OM. Here, we found that CBD alleviated the severity of 5-FU-induced OM in mice, including improved survival, decreased body weight loss, reduced ulcer sizes, and improved clinical scores. Histologically, CBD restored epithelial thickness and normal structure in tongue tissues. Meanwhile, CBD attenuated reactive oxygen species (ROS) overproduction and improved the antioxidant response, suppressed the inflammatory response, promoted the proliferation of epithelial cells, and inhibited 5-FU-induced apoptosis. In vitro, consistent outcomes showed that CBD suppressed cellular ROS levels, enhanced antioxidant ability, reduced inflammatory response, promoted proliferation, and inhibited apoptosis in 5-FU-treated HOK cells. In particular, CBD upregulated the expression levels of antioxidant enzymes, heme oxygenase-1 (HO-1) and NAD(P)H quinine oxidoreductase 1 (NQO1), by increasing the expression and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and decreasing Kelch-like ECH-associated protein 1 (Keap1). Notably, the Nrf2 inhibitor ML385 reversed the protective effect of CBD. Nrf2-siRNA transfection also significantly blunted the antioxidant effect of CBD in in vitro OM model. Collectively, our findings suggested that CBD protected against 5-FU-induced OM injury at least partially via the Nrf2/Keap1/ARE signaling pathways, highlighting the therapeutic prospects of CBD as a novel strategy for chemotherapy-induced OM.
Collapse
|
20
|
Kawata M, Teramura T, Ordoukhanian P, Head SR, Natarajan P, Sundaresan A, Olmer M, Asahara H, Lotz MK. Krüppel-like factor-4 and Krüppel-like factor-2 are important regulators of joint tissue cells and protect against tissue destruction and inflammation in osteoarthritis. Ann Rheum Dis 2022; 81:annrheumdis-2021-221867. [PMID: 35534137 PMCID: PMC9643672 DOI: 10.1136/annrheumdis-2021-221867] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/24/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Analysing expression patterns of Krüppel-like factor (KLF) transcription factors in normal and osteoarthritis (OA) human cartilage, and determining functions and mechanisms of KLF4 and KLF2 in joint homoeostasis and OA pathogenesis. METHODS Experimental approaches included human joint tissues cells, transgenic mice and mouse OA model with viral KLF4 gene delivery to demonstrate therapeutic benefit in structure and pain improvement. Mechanistic studies applied global gene expression analysis and chromatin immunoprecipitation sequencing (ChIP-seq). RESULTS Several KLF genes were significantly decreased in OA cartilage. Among them, KLF4 and KLF2 were strong inducers of cartilage collagen genes and Proteoglycan-4. Cartilage-specific deletion of Klf2 in mature mice aggravated severity of experimental OA. Transduction of human chondrocytes with Adenovirus (Ad) expressing KLF4 or KLF2 enhanced expression of major cartilage extracellular matrix (ECM) genes and SRY-box transcription factor-9, and suppressed mediators of inflammation and ECM-degrading enzymes. Ad-KLF4 and Ad-KLF2 enhanced similar protective functions in meniscus cells and synoviocytes, and promoted chondrocytic differentiation of human mesenchymal stem cells. Viral KLF4 delivery into mouse knees reduced severity of OA-associated changes in cartilage, meniscus and synovium, and improved pain behaviours. ChIP-seq analysis suggested that KLF4 directly bound cartilage signature genes. Ras-related protein-1 signalling was the most enriched pathway in KLF4-transduced cells, and its signalling axis was involved in upregulating cartilage ECM genes by KLF4 and KLF2. CONCLUSIONS KLF4 and KLF2 may be central transcription factors that increase protective and regenerative functions in joint tissue cells, suggesting that KLF gene transfer or molecules upregulating KLFs are therapeutic candidates for OA.
Collapse
Affiliation(s)
- Manabu Kawata
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University, Osaka-Sayama, Osaka, Japan
| | - Philip Ordoukhanian
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Steven R Head
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Padmaja Natarajan
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Aishwarya Sundaresan
- Center for Computational Biology & Bioinformatics and Genomics Core, Scripps Research, La Jolla, California, USA
| | - Merissa Olmer
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Hiroshi Asahara
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Martin K Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
21
|
Rytkönen KT, Faux T, Mahmoudian M, Heinosalo T, Nnamani MC, Perheentupa A, Poutanen M, Elo LL, Wagner GP. Histone H3K4me3 breadth in hypoxia reveals endometrial core functions and stress adaptation linked to endometriosis. iScience 2022; 25:104235. [PMID: 35494227 PMCID: PMC9051620 DOI: 10.1016/j.isci.2022.104235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/11/2022] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Trimethylation of histone H3 at lysine 4 (H3K4me3) is a marker of active promoters. Broad H3K4me3 promoter domains have been associated with cell type identity, but H3K4me3 dynamics upon cellular stress have not been well characterized. We assessed this by exposing endometrial stromal cells to hypoxia, which is a major cellular stress condition. We observed that hypoxia modifies the existing H3K4me3 marks and that promoter H3K4me3 breadth rather than height correlates with transcription. Broad H3K4me3 domains mark genes for endometrial core functions and are maintained or selectively extended upon hypoxia. Hypoxic extension of H3K4me3 breadth associates with stress adaptation genes relevant for the survival of endometrial cells including transcription factor KLF4, for which we found increased protein expression in the stroma of endometriosis lesions. These results substantiate the view on broad H3K4me3 as a marker of cell identity genes and reveal participation of H3K4me3 extension in cellular stress adaptation.
Collapse
Affiliation(s)
- Kalle T. Rytkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
- Yale Systems Biology Institute, West Haven, CT 06516, USA
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20014 Turku, Finland
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA
- Corresponding author
| | - Thomas Faux
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Mehrad Mahmoudian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20014 Turku, Finland
| | - Taija Heinosalo
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20014 Turku, Finland
| | - Mauris C. Nnamani
- Yale Systems Biology Institute, West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA
| | - Antti Perheentupa
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20014 Turku, Finland
- Department of Obstetrics and Gynecology, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, 20014 Turku, Finland
| | - Laura L. Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20014 Turku, Finland
| | - Günter P. Wagner
- Yale Systems Biology Institute, West Haven, CT 06516, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06511, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale Medical School, New Haven, CT 06510, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Corresponding author
| |
Collapse
|
22
|
Methylene blue prevents osteoarthritis progression and relieves pain in rats via upregulation of Nrf2/PRDX1. Acta Pharmacol Sin 2022; 43:417-428. [PMID: 33833406 DOI: 10.1038/s41401-021-00646-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress-related cartilage degeneration, synovitis, and joint pain play vital roles in the progress of osteoarthritis (OA). Anti-oxidative stress agents not only prevent structural damage progression but also relieve OA-related pain. In this study, we investigated the therapeutic effect of methylene blue (MB), a classical and important anti-oxidant with strong neural affinity. Experimental OA was established in rats by radial transection of medial collateral ligament and medial meniscus (MCLT + MMT) of the right knee joint. The OA rats received intra-articular injection of MB (1 mg/kg) every week starting one week after surgery. We showed that MB administration exerted significant cartilage protection, synovitis inhibition as well as pain relief in OA rats. In human chondrocytes and fibroblast-like synoviocytes, MB significantly attenuated tert-butyl hydroperoxide (TBHP)-induced inflammatory response and oxidative stress. We demonstrated that these effects of MB resulted from dual targets of important antioxidant enzymes, Nrf2 and PRDX1, which also mutually reinforcing and participated in an interaction. Furthermore, we found that calcitonin gene-related peptide (CGRP), a neural inflammatory mediator, was accumulated around the vessel in synovium and subchondral bone in OA rats and in TBHP-treated primary cortical neurons; MB administration significantly inhibited CGRP expression through upregulation of Nrf2 and PRDX1. Taken together, these results suggest that MB ameliorates oxidative stress via Nrf2/PRDX1 regulation to prevent progression and relieve pain of OA.
Collapse
|
23
|
Wang D, Fan Y, Ma J, Gao C, Liu X, Zhao Z, Wei H, Yang G, Huang J, Jiang R, Zhang J. Atorvastatin combined with dexamethasone promote hematoma absorption in an optimized rat model of chronic subdural hematoma. Aging (Albany NY) 2021; 13:24815-24828. [PMID: 34813498 PMCID: PMC8660610 DOI: 10.18632/aging.203717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022]
Abstract
Previous studies found that atorvastatin and dexamethasone were effective in promoting the absorption of chronic subdural hematoma. In this study, we aimed to investigate the effect of pharmacotherapy in an optimized rat model of chronic subdural hematoma. Rat model of chronic subdural hematoma via a bEnd.3 cell and Matrigel mix was established and dynamic changes in different drug treatment groups were tested. The hematoma gradually increased, peaked on the fifth day (263.8±52.85 μl), and was completely absorbed in two weeks. Notably, Kruppelle-like factor 2 expression was significantly decreased with increasing hematoma volume, and then increased in the repair period. The expression of IL-10 was increased and peaked on 7 days, and then decreased at 14 days. The dynamic trends of IL-6, IL-8, MMP-9, and VEGF were also increased first and then decreased. Both monotherapy and the combined treatment by atorvastatin and dexamethasone could counteract the inflammatory activities, decrease hematoma permeability, and improve hematoma absorption, however, most prominent in combined group. The combined treatment could more effectively increase Kruppelle-like factor 2 and ZO-1 expression, attenuate the expression of NF-κb. Most importantly, the combined treatment enhanced the neural functional prognosis and reduced the mortality of chronic subdural hematoma rats. According to our results, the combined treatment could more effectively attenuate inflammatory. And it could also enhance angiogenic activities which could promote the stability of local function and structure of the hematoma cavity, reduce the hematoma volume and improve the outcomes of rats with chronic subdural hematoma than single treatments in the optimized chronic subdural hematoma model.
Collapse
Affiliation(s)
- Dong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Yueshan Fan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China.,Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Jun Ma
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Chuang Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Xuanhui Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China.,Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Zilong Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Huijie Wei
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Guili Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Jinhao Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, Tianjin, China
| |
Collapse
|
24
|
Chen Y, Sun Y, Xu Y, Lin WW, Luo Z, Han Z, Liu S, Qi B, Sun C, Go K, Kang XR, Chen J. Single-Cell Integration Analysis of Heterotopic Ossification and Fibrocartilage Developmental Lineage: Endoplasmic Reticulum Stress Effector Xbp1 Transcriptionally Regulates the Notch Signaling Pathway to Mediate Fibrocartilage Differentiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7663366. [PMID: 34737845 PMCID: PMC8563124 DOI: 10.1155/2021/7663366] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/21/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Regeneration of fibrochondrocytes is essential for the healing of the tendon-bone interface (TBI), which is similar to the formation of neurogenic heterotopic ossification (HO). Through single-cell integrative analysis, this study explored the homogeneity of HO cells and fibrochondrocytes. METHODS This study integrated six datasets, namely, GSE94683, GSE144306, GSE168153, GSE138515, GSE102929, and GSE110993. The differentiation trajectory and key transcription factors (TFs) for HO occurrence were systematically analyzed by integrating single-cell RNA (scRNA) sequencing, bulk RNA sequencing, and assay of transposase accessible chromatin seq. The differential expression and enrichment pathways of TFs in heterotopically ossified tissues were identified. RESULTS HO that mimicked pathological cells was classified into HO1 and HO2 cell subsets. Results of the pseudo-temporal sequence analysis suggested that HO2 is a differentiated precursor cell of HO1. The analysis of integrated scRNA data revealed that ectopically ossified cells have similar transcriptional characteristics to cells in the fibrocartilaginous zone of tendons. The modified SCENIC method was used to identify specific transcriptional regulators associated with ectopic ossification. Xbp1 was defined as a common key transcriptional regulator of ectopically ossified tissues and the fibrocartilaginous zone of tendons. Subsequently, the CellPhoneDB database was completed for the cellular ligand-receptor analysis. With further pathway screening, this study is the first to propose that Xbp1 may upregulate the Notch signaling pathway through Jag1 transcription. Twenty-four microRNAs were screened and were found to be potentially associated with upregulation of XBP1 expression after acute ischemic stroke. CONCLUSION A systematic analysis of the differentiation landscape and cellular homogeneity facilitated a molecular understanding of the phenotypic similarities between cells in the fibrocartilaginous region of tendon and HO cells. Furthermore, by identifying Xbp1 as a hub regulator and by conducting a ligand-receptor analysis, we propose a potential Xbp1/Jag1/Notch signaling pathway.
Collapse
Affiliation(s)
- Yisheng Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province 271000, China
| | - Wei-Wei Lin
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009 Zhejiang, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhihua Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Shaohua Liu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beijie Qi
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chenyu Sun
- Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, 60657 Illinois, USA
| | - Ken Go
- Department of Clinical Training Centre, St. Marianna Hospital, Tokyo, Japan
| | - x.-R. Kang
- Shanghai Jiao Tong University, Shanghai 200080, China
| | - Jiwu Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| |
Collapse
|
25
|
Li Z, Wang J, Ma Y. Montelukast attenuates interleukin IL-1β-induced oxidative stress and apoptosis in chondrocytes by inhibiting CYSLTR1 (Cysteinyl Leukotriene Receptor 1) and activating KLF2 (Kruppel Like Factor 2). Bioengineered 2021; 12:8476-8484. [PMID: 34565285 PMCID: PMC8806840 DOI: 10.1080/21655979.2021.1984003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Montelukast is a cysteinyl leukotriene receptor 1 (CysLTR1) antagonist widely used to suppress the inflammatory response in asthma and allergic rhinitis. This study aimed to investigate the potential impacts of montelukast on osteoarthritis (OA) progression. To determine the role of montelukast in OA, the expression of CysLTR1 was first examined by quantitative reverse transcription PCR (RT-qPCR) and western blot in IL-1β-induced ATDC5 cells treated with or without montelukast. Subsequently, the impacts of montelukast on cell viability and oxidative stress were measured by Cell-Counting-Kit-8 (CCK-8), commercial kits and western blot. Oxidative stress-related protein expressions were determined by western blot analysis in Il-1β-induced ATDC5 cells. Cell apoptosis and cartilage degradation were examined by TdT-mediated dUTP Nick-End Labeling (TUNEL) assay, western blot and RT-qPCR. KLF2 expression was measured in IL-1β-induced ATDC5 cells treated with montelukast. After interference with small interfering RNA (siRNA)-KLF2 in ATDC5 cells, the loss-of-function assays were also performed in same ways. CysLTR1 expression was elevated in IL-1β-induced ATDC5 cells but inhibited significantly by montelukast. Montelukast attenuated the oxidative stress and apoptosis, improved cell viability. Moreover, montelukast enhanced KLF2 expression. After transfected with siRNA-KLF2, montelukast attenuated cell injury, oxidative stress, apoptosis and cartilage degradation in IL-1β-induced ATDC5 cells by activating KLF2.In summary, this work elaborates the evidence that montelukast could attenuate oxidative stress and apoptosis in IL-1β-induced chondrocytes by inhibiting CysLTR1 and activating KLF2, which can guide the therapeutic strategies of montelukast for OA development in the future.
Collapse
Affiliation(s)
- Zongwei Li
- School of Pharmaceutical Engineering, Guangdong Food and Drug Vocational College, Guangzhou City, Guangdong Province, China
| | - Jianming Wang
- School of Pharmaceutical Engineering, Guangdong Food and Drug Vocational College, Guangzhou City, Guangdong Province, China
| | - Yumin Ma
- Department of Pharmaceutical Machinery, Maternal and Child Health and Family Planning Technical Service Center, Wuwei City, Gansu Province, China
| |
Collapse
|
26
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China ,grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Peng Liu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
27
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z+10.1038/s41413-021-00163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2024] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
28
|
Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
|
29
|
Hou M, Zhang Y, Zhou X, Liu T, Yang H, Chen X, He F, Zhu X. Kartogenin prevents cartilage degradation and alleviates osteoarthritis progression in mice via the miR-146a/NRF2 axis. Cell Death Dis 2021; 12:483. [PMID: 33986262 PMCID: PMC8119954 DOI: 10.1038/s41419-021-03765-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common articular degenerative disease characterized by loss of cartilage matrix and subchondral bone sclerosis. Kartogenin (KGN) has been reported to improve chondrogenic differentiation of mesenchymal stem cells. However, the therapeutic effect of KGN on OA-induced cartilage degeneration was still unclear. This study aimed to explore the protective effects and underlying mechanisms of KGN on articular cartilage degradation using mice with post-traumatic OA. To mimic the in vivo arthritic environment, in vitro cultured chondrocytes were exposed to interleukin-1β (IL-1β). We found that KGN barely affected the cell proliferation of chondrocytes; however, KGN significantly enhanced the synthesis of cartilage matrix components such as type II collagen and aggrecan in a dose-dependent manner. Meanwhile, KGN markedly suppressed the expression of matrix degradation enzymes such as MMP13 and ADAMTS5. In vivo experiments showed that intra-articular administration of KGN ameliorated cartilage degeneration and inhibited subchondral bone sclerosis in an experimental OA mouse model. Molecular biology experiments revealed that KGN modulated intracellular reactive oxygen species in IL-1β-stimulated chondrocytes by up-regulating nuclear factor erythroid 2-related factor 2 (NRF2), while barely affecting its mRNA expression. Microarray analysis further revealed that IL-1β significantly up-regulated miR-146a that played a critical role in regulating the protein levels of NRF2. KGN treatment showed a strong inhibitory effect on the expression of miR-146a in IL-1β-stimulated chondrocytes. Over-expression of miR-146a abolished the anti-arthritic effects of KGN not only by down-regulating the protein levels of NRF2 but also by up-regulating the expression of matrix degradation enzymes. Our findings demonstrate, for the first time, that KGN exerts anti-arthritic effects via activation of the miR-146a-NRF2 axis and KGN is a promising heterocyclic molecule to prevent OA-induced cartilage degeneration.
Collapse
Affiliation(s)
- Mingzhuang Hou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Yijian Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Xinfeng Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Xi Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China.
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China. .,Orthopaedic Institute, Medical College, Soochow University, Suzhou, China.
| |
Collapse
|
30
|
Sanada Y, Tan SJO, Adachi N, Miyaki S. Pharmacological Targeting of Heme Oxygenase-1 in Osteoarthritis. Antioxidants (Basel) 2021; 10:antiox10030419. [PMID: 33803317 PMCID: PMC8001640 DOI: 10.3390/antiox10030419] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) is a common aging-associated disease that clinically manifests as joint pain, mobility limitations, and compromised quality of life. Today, OA treatment is limited to pain management and joint arthroplasty at the later stages of disease progression. OA pathogenesis is predominantly mediated by oxidative damage to joint cartilage extracellular matrix and local cells such as chondrocytes, osteoclasts, osteoblasts, and synovial fibroblasts. Under normal conditions, cells prevent the accumulation of reactive oxygen species (ROS) under oxidatively stressful conditions through their adaptive cytoprotective mechanisms. Heme oxygenase-1 (HO-1) is an iron-dependent cytoprotective enzyme that functions as the inducible form of HO. HO-1 and its metabolites carbon monoxide and biliverdin contribute towards the maintenance of redox homeostasis. HO-1 expression is primarily regulated at the transcriptional level through transcriptional factor nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2), specificity protein 1 (Sp1), transcriptional repressor BTB-and-CNC homology 1 (Bach1), and epigenetic regulation. Several studies report that HO-1 expression can be regulated using various antioxidative factors and chemical compounds, suggesting therapeutic implications in OA pathogenesis as well as in the wider context of joint disease. Here, we review the protective role of HO-1 in OA with a focus on the regulatory mechanisms that mediate HO-1 activity.
Collapse
Affiliation(s)
- Yohei Sanada
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 7348551, Japan;
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
| | - Sho Joseph Ozaki Tan
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
| | - Shigeru Miyaki
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 7348551, Japan;
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
- Correspondence: ; Tel.: +81-82-257-5231
| |
Collapse
|
31
|
Fan YS, Wang B, Wang D, Xu X, Gao C, Li Y, Zhang S, Yang GL, Liu X, Jiang RC, Zhang JN. Atorvastatin combined with low-dose dexamethasone for vascular endothelial cell dysfunction induced by chronic subdural hematoma. Neural Regen Res 2021; 16:523-530. [PMID: 32985481 PMCID: PMC7996011 DOI: 10.4103/1673-5374.293152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Atorvastatin has been shown to be a safe and effective non-surgical treatment option for patients with chronic subdural hematoma. However, treatment with atorvastatin is not effective in some patients, who must undergo further surgical treatment. Dexamethasone has anti-inflammatory and immunomodulatory effects, and low dosages are safe and effective for the treatment of many diseases, such as ankylosing spondylitis and community-acquired pneumonia. However, the effects of atorvastatin and low-dose dexamethasone for the treatment of chronic subdural hematoma remain poorly understood. Hematoma samples of patients with chronic subdural hematoma admitted to the General Hospital of Tianjin Medical University of China were collected and diluted in endothelial cell medium at 1:1 as the hematoma group. Atorvastatin, dexamethasone, or their combination was added to the culture medium. The main results were as follows: hopping probe ion conductance microscopy and permeability detection revealed that the best dosages to improve endothelial cell permeability were 0.1 μM atorvastatin and 0.1 μM dexamethasone. Atorvastatin, dexamethasone, or their combination could markedly improve the recovery of injured endothelial cells. Mice subcutaneously injected with diluted hematoma solution and then treated with atorvastatin, dexamethasone, or their combination exhibited varying levels of rescue of endothelial cell function. Hopping probe ion conductance microscopy, western blot assay, and polymerase chain reaction to evaluate the status of human cerebral endothelial cell status and expression level of tight junction protein indicated that atorvastatin, dexamethasone, or their combination could reduce subcutaneous vascular leakage caused by hematoma fluid. Moreover, the curative effect of the combined treatment was significantly better than that of either single treatment. Expression of Krüppel-like factor 2 protein in human cerebral endothelial cells was significantly increased, as was expression of the tight junction protein and vascular permeability marker vascular endothelial cadherin in each treatment group compared with the hematoma stimulation group. Hematoma fluid in patients with chronic subdural hematoma may damage vascular endothelial cells. However, atorvastatin combined with low-dose dexamethasone could rescue endothelial cell dysfunction by increasing the expression of tight junction proteins after hematoma injury. The effect of combining atorvastatin with low-dose dexamethasone was better than that of atorvastatin alone. Increased expression of Krüppel-like factor 2 may play an important role in the treatment of chronic subdural hematoma. The animal protocols were approved by the Animal Care and Use Committee of Tianjin Medical University of China on July 31, 2016 (approval No. IRB2016-YX-036). The study regarding human hematoma samples was approved by the Ethics Committee of Tianjin Medical University of China on July 31, 2018 (approval No. IRB2018-088-01).
Collapse
Affiliation(s)
- Yue-Shan Fan
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City; Graduate School, Tianjin Medical University, Tianjin, China
| | - Bo Wang
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City; Graduate School, Tianjin Medical University, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City; Graduate School, Tianjin Medical University, Tianjin, China
| | - Xin Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chuang Gao
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Ying Li
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Shu Zhang
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Gui-Li Yang
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xiao Liu
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Rong-Cai Jiang
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Jian-Ning Zhang
- Department of Neurosurgery, General Hospital of Tianjin Medical University; Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| |
Collapse
|
32
|
Xiao P, Zhu X, Sun J, Zhang Y, Qiu W, Li J, Wu X. MicroRNA-613 alleviates IL-1β-induced injury in chondrogenic CHON-001 cells by targeting fibronectin 1. Am J Transl Res 2020; 12:5308-5319. [PMID: 33042421 PMCID: PMC7540165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is an aging-related chronic degenerative joint disease. A number of miRNAs have been found to be involved in the development of OA, but the role of miR-613 in OA remains unclear. Thus, this study aimed to investigate the role of miR-613 during the progression of OA. METHODS CHON-001 cells were transfected with miR-613 agonist for 48 h, and then exposed to 10 ng/mL IL-1β for 24 h. Cell viability, cell proliferation and cell apoptosis in CHON-001 cells were assessed by CCK-8, immunofluorescence, and flow cytometry assays, respectively. In addition, the dual luciferase reporter system assay was used to determine the interaction of miR-613 and fibronectin 1 in CHON-001 cells. RESULTS The level of miR-613 was significantly decreased in IL-1β-treated CHON-001 cells. Overexpression of miR-613 markedly inhibited IL-1β-induced apoptosis in CHON-001 cells. In addition, upregulation of miR-613 obviously alleviated IL-1β-induced inflammatory response and cartilage matrix degradation in CHON-001 cells. Meanwhile, fibronectin 1 was identified as a direct binding target of miR-613 in CHON-001 cells. Overexpression of miR-613 alleviated IL-1β-induced injury in CHON-001 cells via downregulating the expression of fibronectin 1. Furthermore, overexpression of miR-613 alleviated cartilage degradation, and reduced OARSI scores and subchondral bone thickness in a mouse model of OA. CONCLUSION Our data indicated that overexpression of miR-613 could inhibit IL-1β-induced injury in CHON-001 cells via decreasing the level fibronectin 1 in vitro, and alleviate the symptoms of OA in vivo. Therefore, miR-613 might be a potential therapeutic option for the treatment of OA.
Collapse
Affiliation(s)
- Peng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Xu Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Jinpeng Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Yuhang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Weijian Qiu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Jianqiang Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| | - Xuejian Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University Zhengzhou 450000, Henan, P. R. China
| |
Collapse
|
33
|
Zhao J, Su Y, Jiao J, Wang Z, Fang X, He X, Zhang X, Liu Z, Xu X. Identification of lncRNA and mRNA Biomarkers in Osteoarthritic Degenerative Meniscus by Weighted Gene Coexpression Network and Competing Endogenous RNA Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2123787. [PMID: 32685450 PMCID: PMC7341399 DOI: 10.1155/2020/2123787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/26/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) play a crucial role in varieties of biological processes. This study is aimed at investigating meniscal degeneration-specific lncRNAs and mRNAs and their related networks in knee osteoarthritis (KOA). METHODS The dataset GSE98918, which included 24 meniscus samples and related clinical data, was downloaded from the Gene Expression Omnibus database. The differentially expressed lncRNAs and mRNAs in the meniscus between KOA and control groups were identified. Based on the enriched differentially expressed lncRNAs and mRNAs, we constructed the coexpression network using WGCNA (weighted correlation network analysis) and identified the critical module related to KOA. For mRNAs in the key module, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were carried out using the DAVID database. A competing endogenous RNA network (ceRNA) based on the screened mRNAs, lncRNAs, and related miRNAs was constructed to reveal presumptive biomarkers further. Finally, the hub lncRNAs and mRNAs were screened, and the diagnostic value was evaluated using a receiver operating characteristic (ROC) curve. Hub mRNAs were validated using the dataset GSE113825. RESULTS We screened 208 significantly differentially expressed lncRNAs and mRNAs in menisci between the KOA and non-KOA samples, which were enriched in sixteen modules using WGCNA, especially the green module. Coexpression network based on the enriched differentially expressed lncRNAs and mRNAs in the green module uncovered 5 lncRNAs and 56 mRNAs. The lncRNA-miRNA-mRNA ceRNA network revealed that lnc-HLA-DQA1-5, lnc-RP11-127H5.1.1-1, lnc-RTN2-1, IGFBP4 (insulin-like growth factor binding protein 4), and KLF2 (Kruppel-like factor 2) were significantly correlated with the meniscus degeneration of KOA. ROC curve analysis revealed that these hub lncRNAs and mRNAs showed excellent diagnostic value for KOA. CONCLUSIONS These hub lncRNAs and mRNAs were potential prognostic biomarkers for the meniscus degeneration of KOA. Further studies are required to validate these new biomarkers and better understand the pathological process of the meniscus degeneration of KOA.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Yu Su
- Harbin Fifth Hospital, Jiankang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Jianfei Jiao
- Harbin Fifth Hospital, Jiankang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Zhengchun Wang
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xiangchun Fang
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xuefeng He
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xiaofeng Zhang
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Zhao Liu
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xilin Xu
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| |
Collapse
|