1
|
Wang F, Li J, Zhang Z, Huang G, Zhang X, Liu Q, Xiao W, Liu F, Sun J, Liu Y, Ma Y, Zhuang R, Du Y, Wang X, Gao C, Gu X. Baicalin reduced vandetanib induced myocardial injury by regulating redox balance and NLRP3 inflammasome pathway. Tissue Cell 2025; 94:102795. [PMID: 39987774 DOI: 10.1016/j.tice.2025.102795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025]
Abstract
Baicalin has garnered attention for its potential therapeutic effects on various cardiovascular conditions, including drug-induced cardiac injury. In this study, we utilized a murine model to explore the protective role of baicalin against cardiac dysfunction induced by vandetanib. Our findings indicate that baicalin administration effectively ameliorated vandetanib-induced cardiac injury. Echocardiographic assessments revealed significant improvements in the myocardial contraction in mice treated with baicalin compared with those receiving vandetanib alone. Histological analysis revealed reduced myocardial inflammation and fibrosis in baicalin-treated mice. Specifically, baicalin suppressed proinflammatory factors such as IL-6, IL-1β, and TNF-α, thereby attenuating the inflammatory response triggered by vandetanib. Moreover, baicalin inhibited myocardial apoptosis, as evidenced by decreased levels of Caspase-3, Bax, and p53, while concurrently elevated expression of the antiapoptotic protein Bcl-2. Mechanistically, baicalin-mediated inhibition of the NLRP3 inflammasome pathway has emerged as a crucial aspect of its cardioprotective action and promotes redox balance in myocardial cells under vandetanib-induced oxidative stress. It upregulated the expression of the antioxidant enzymes SOD1 and SOD2, thereby mitigating intracellular ROS accumulation and preserving cardiomyocyte viability. In conclusion, our study highlights baicalin as a promising therapeutic agent for mitigating vandetanib-induced cardiac injury through multiple mechanisms, including anti-inflammatory, antiapoptotic, antioxidant, and NLRP3 inflammasome inhibitory actions. Our findings will be further validated in clinical trials and explore the translational potential of baicalin in treating drug-induced cardiotoxicity in humans.
Collapse
Affiliation(s)
- Fen Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Jianwei Li
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Zhixuan Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Guangyi Huang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Xiaodong Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Qian Liu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Wang Xiao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Fengqi Liu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Jialong Sun
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Yankui Liu
- Department of Pathology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Yiyi Ma
- Department of Ultrasound Medicine,The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Ruijuan Zhuang
- Department of Geriatric Medicine, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China
| | - Yingqiang Du
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University; Suzhou Municipal Hospital; Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215008, China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China.
| | - Changzheng Gao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China.
| | - Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214000, China.
| |
Collapse
|
2
|
Li B, Wang K, Wang X, Zhang Z, Huang G, Ma Y, Du Y, Gu X, Hui J. p16 INK4a Aggravated Sepsis-associated Cardiac Injury by Inhibiting the PI3K/AKT Pathway and Inducing Redox Imbalance. J Cardiovasc Transl Res 2025; 18:375-391. [PMID: 39809973 DOI: 10.1007/s12265-024-10588-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
Severe sepsis can promote myocardial injury and cardiac dysfunction, but role of p16 in sepsis-induced myocardial injury remains undefined. PBMCs were collected from patients. Expression of inflammatory factors and NLRP3 pathway were detected by Western blotting and qPCR in WT and p16KO mice. Then detect cardiomyocyte apoptosis and ROS levels in vitro. Detailed pathways and mechanisms were revealed through quantitative proteomic analysis combined with GSEA and KEGG analysis. p16 was overexpressed in PBMCs of patient. p16 knockout alleviated cardiac dysfunction in LPS-induced mice and inhibited NLRP3 inflammasome pathway in vivo and in vitro. Quantitative proteomic analysis revealed that p16 knockout contributed to the activation of the PI3K/Akt pathway in LPS-induced cardiac injury. p16 knockout promoted activation of the PI3K/Akt pathway and ameliorated NLRP3 pathway inhibition and redox imbalance thus improving cardiac function in LPS-induced cardiomyopathy mice.
Collapse
Affiliation(s)
- Baihong Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University Suzhou, Jiangsu, 215000, China
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Kai Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Zhixuan Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Guangyi Huang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Yiyi Ma
- Department of Ultrasonography, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Yingqiang Du
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215008, Jiangsu, China.
| | - Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China.
| | - Jie Hui
- Department of Cardiology, The First Affiliated Hospital of Soochow University Suzhou, Jiangsu, 215000, China.
| |
Collapse
|
3
|
Wei YF, Zhang HL, Li LZ, Lv Y, Li H, Li Z, Yu FL, Jiang T, Zhang TY, Xin F, Ma C, Ren YX. Sirt1 blocks nucleus pulposus and macrophages crosstalk by inhibiting RelA/Lipocalin 2 axis. J Orthop Translat 2025; 50:30-43. [PMID: 39758288 PMCID: PMC11699611 DOI: 10.1016/j.jot.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Background Intervertebral disc degeneration (IVDD) stands as a primary pathophysiological driver of low back pain, yet no therapeutic intervention effectively arrests its progression. Evidence shows that certain Sirt1 agonists may confer protective effects on intervertebral discs, but the underlying mechanisms remain unclear. This study aims to delineate the interaction between Sirt1 and the inflammatory microenvironment, offering potential novel avenues for IVDD prevention and treatment. Methods In vitro IL-1β-induced nucleus pulposus cells (NPCs) degenerative model and in vivo a mouse annulus fibrosus needle puncture model in Sirt1 transgenic (Sirt1TG) and the same litter WT mice were used to investigate the role of Sirt1 in homeostasis and inflammation. Mechanistic insights were obtained through RNA sequencing, co-immunoprecipitation (Co-IP), luciferase assays, and chromatin immunoprecipitation-(ChIP)-PCR. A co-culture system of Raw264.7 and NPCs was employed to assess the involvement of Lipocalin 2. Results Our study demonstrated reduced Sirt1 expression in degenerating human nucleus pulposus (NP) tissue. Both in vitro and in vivo data revealed that NP-specific overexpression of Sirt1 inhibited extracellular matrix degradation and inflammation. Mechanistically, Sirt1 suppressed the acetylation of RelA/p65 at lysine 310 and phosphorylation at serine 536, with the C-terminus of Sirt1 and the RHD-NLS domain of RelA mediating to their interaction. Furthermore, NPCs-derived Lipocalin 2 was identified as a cytokine involved in macrophage chemotaxis and M1 polarization to exacerbate inflammation. Conclusion Our work revealed that Sirt1 negatively regulates Lipocalin 2, thereby ameliorating the inflammatory milieu and blocking NPCs and macrophages crosstalk. The Translational Potential of this Article This study illuminates the crucial role and molecular mechanisms of Sirt1 in regulating the NP microenvironment. These insights shed light on strategies for the prevention and treatment of IVDD-related herniation and low back pain. By pinpointing specific biological targets, the screening of smallmolecule compounds with significant clinical implications can be facilitated. This translational innovation promises to optimize cells communication within intervertebral disc microenvironment via localized drug delivery, potentially improving patient outcomes and satisfaction following spinal fusion or discectomy surgeries.
Collapse
Affiliation(s)
- Yi-Fan Wei
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - He-Long Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Ling-Zhi Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - You Lv
- Department of Orthopaedics, Lianyungang Clinical College of Nanjing Medical University, 6 Zhenhua East Rd, Lianyungang, 221000, China
| | - He Li
- Department of Sports Medicine, Lianyungang Clinical College of Nanjing Medical University, 6 Zhenhua East Rd, Lianyungang, 221000, China
| | - Zhi Li
- Department of Orthopaedics, Geriatric Hospital of Nanjing Medical University, 65 Jiangsu Rd, Nanjing, 210024, China
| | - Feng-Lei Yu
- Department of Trauma and Orthopaedics, The First People's Hospital of Kunshan, 566 East Qianjin Rd, Suzhou, 215000, China
| | - Tao Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Tian-You Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Feng Xin
- Department of Orthopaedics, Xuzhou Cancer Hospital, 131 Huancheng Rd, Xuzhou, 221005, China
| | - Cheng Ma
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| | - Yong-Xin Ren
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029, China
| |
Collapse
|
4
|
Cui X, Xu F, Pang X, Fan C, Jiang H. WTAP-Mediated m6A Modification of TRAIL-DR4 Suppresses MH7A Cell Apoptosis. Int J Rheum Dis 2025; 28:e70065. [PMID: 39797510 DOI: 10.1111/1756-185x.70065] [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: 10/09/2024] [Revised: 12/17/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND N6-methyladenosine (m6A) is one of the most conserved internal RNA modifications, which has been implicated in many biological processes, such as apoptosis and proliferation. Wilms tumor 1-associating protein (WTAP), as a key component of m6A methylation, is a nuclear protein that has been associated with the regulation of proliferation and apoptosis. Rheumatoid arthritis (RA), a systemic, infiltrating autoimmune disease, is characterized by synovial hyperplasia. However, little is known about the precise role of WTAP in RA. This study investigated the role of the WTAP-mediated m6A modification of TNF-related apoptosis-inducing ligand death receptor 4 (TRAIL-DR4) in RA. METHOD Methyltransferase WTAP overexpression plasmids and small interfering RNAs were constructed and transfected into MH7A cells. Immunofluorescence (IF) staining, quantitative reverse transcription polymerase chain reaction (RT-qPCR), and Western blot were used to detect changes in the expression of WTAP, the B-cell lymphoma 2 (BCL2) gene family, BCL2-associated X (BAX) and TRAIL-DR4 expression, and the effects of WTAP overexpression on cell viability, cell cycle, apoptosis, and proliferation were assessed by a cell counting kit-8 (CCK-8), flow cytometry, and transmission electron microscopy (TEM). The m6A modification of TRAIL-DR4 was verified by m6A methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) and its stability was assessed by an actinomycin D assay. RESULTS Overexpression of WTAP not only increased the levels of WTAP and BCL2, and decreased the levels of BAX and TRAIL-DR4, but also significantly inhibited MH7A cell apoptosis and promoted cell viability and proliferation, while WTAP silencing led to the opposite trend. The SRAMP online database predicted that TRAIL-DR4 has multiple potential methylation-binding sites, and fluorescence in situ hybridization (FISH) combined with IF showed that WTAP and TRAIL-DR4 were mainly expressed in both the nucleus and cytoplasm. MeRIP-qPCR and actinomycin D analysis experiments revealed that WTAP could promote the m6A level of TRAIL-DR4, decrease the stability of TRAIL-DR4 mRNA, and subsequently inhibit apoptosis. CONCLUSION This study suggests that WTAP-mediated m6A modification of TRAIL-DR4 suppresses MH7A cell apoptosis. This discovery offers a new focus and avenue for the clinical treatment of RA, while also extending our understanding of the pathophysiology of RA from the standpoint of m6A alteration.
Collapse
Affiliation(s)
- Xiaoya Cui
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Fengxia Xu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xue Pang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chang Fan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hui Jiang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
5
|
Zhang Q, He X, Chen W, Jiu J, Gao C, Gao T. Vitamin D3 attenuates autoimmune thyroiditis by regulating Th17/Treg cell differentiation via YAP/JAK1/STAT1 axis. Immunol Lett 2024; 269:106890. [PMID: 38959983 DOI: 10.1016/j.imlet.2024.106890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Autoimmune thyroiditis (AITD) is an organ-specific autoimmune disease. Substantial evidence suggests that Vitamin D (VitD) deficiency is closely associated with an increased risk of AITD. However, the effects of VitD3 on immune cells, especially Th17/Treg cell subsets, and the underlying molecular mechanism in AITD have not yet been investigated. METHODS An experimental autoimmune thyroiditis (EAT) mouse model was established with a high-iodine diet. After 8 weeks, thyroid injury was assessed using hematoxylin and eosin (H&E) staining. ELISA was employed to measure serum levels of thyroxine (T3 and T4), thyroid autoimmune antibodies (Tg-Ab and TPO-Ab), and inflammatory cytokines. Flow cytometry and multiplex fluorescence immunohistochemical (mIHC) assays were used to analyze Th17/Treg cell subsets. The CCK-8 and flow cytometry assays were used to determine cell viability and apoptosis. RESULTS Administration of VitD3 reduced thyroid follicle destruction, decreased lymphocyte infiltration, and lowered T3, T4, Tg-Ab, and TPO-Ab serum levels in EAT mice. VitD3 treatment also reduced the frequency of Th17 cells while promoting the Treg cell subset both in the thyroid tissue and in the splenocytes cultured in vitro. Furthermore, VitD3 administration suppressed the production of inflammatory cytokines in EAT mice. VitD3 was also found to regulate Treg cells' differentiation, viability, and apoptosis. Mechanistically, we discovered that VitD3 treatment upregulated YAP expression and activated the JAK/STAT pathway. Rescue assays confirmed that depletion of YAP counteracted the effects of VitD3 on Treg cell differentiation and function. CONCLUSION Vitamin D3 attenuates AITD by modulating Th17/Treg cell balance via regulating the YAP/JAK1/STAT1 axis.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Immunology, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, Liaoning 110847, China
| | - Xin He
- Department of Cardiology, First Affiliated Hospital of Jinzhou Medical University, No.40 Songpo Road, Linghe District, Jinzhou, Liaoning 121001, China
| | - Wei Chen
- Department of Immunology, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, Liaoning 110847, China
| | - Junlong Jiu
- Department of Immunology, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, Liaoning 110847, China
| | - Chenghan Gao
- Department of Immunology, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, Liaoning 110847, China
| | - Tianshu Gao
- Department of Immunology, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Huanggu District, Shenyang, Liaoning 110847, China.
| |
Collapse
|
6
|
Tang T, Lu T, Li B, Zhou Y, Huang L, Zhang L, Chen Y, Li H. Deletion of vitamin D receptor exacerbated temporomandibular joint pathological changes under abnormal mechanical stimulation. Life Sci 2024; 353:122913. [PMID: 39004274 DOI: 10.1016/j.lfs.2024.122913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
AIMS Temporomandibular disorder can cause degenerative pathological changes by aseptic inflammation in the temporomandibular joint (TMJ). Vitamin D (VD) is known for maintaining calcium homeostasis, and recent studies indicated that VD and the vitamin D receptor (VDR) are important in inflammatory responses. In this study, we explored the anti-inflammatory effect of VD-VDR signaling axis in TMJ pathological degeneration. MAIN METHODS Mice ablated for Vdr (Vdr-/-res) were fed with a rescue diet to avoid hypocalcemia. With abnormal mechanical stimulation, unilateral anterior crossbite (UAC) induced temporomandibular disorders in mice. Histological staining, immunohistochemistry staining, and micro-CT analysis were performed to evaluate TMJ pathological changes. To identify the mechanisms in the aseptic inflammatory process, in vitro experiments were conducted on wild-type (WT) and Vdr-/- chondrocytes with compressive mechanical stress loading, and the related inflammatory markers were examined. KEY FINDINGS Vdr-/-res mice did not develop rickets with a high calcium rescue diet. The TMJ cartilage thickness in Vdr-/-res mice was significantly decreased with mechanical stress stimulation compared to WT mice. UAC-induced bone resorption was obvious, and the number of osteoclasts significantly increased in Vdr-/-res mice. The proliferation was inhibited and the gene expression of Il1b, Mmp3, and Mmp13 was significantly increased in Vdr-/- chondrocytes. However, WT chondrocytes showed significantly increased Tnfa gene expression as a response to mechanical stress but not in Vdr-/- chondrocytes. SIGNIFICANCE VD-VDR is crucial in TMJ pathological changes under abnormal mechanical stimulation. Deletion of Vdr exacerbated inflammatory response excluding TNFα, inhibited chondrocyte proliferation, and promoted bone resorption in TMJ.
Collapse
Affiliation(s)
- Tianyi Tang
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China; Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, London NW3 2PF, UK.
| | - Tong Lu
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Baochao Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Yiwen Zhou
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China.
| | - Ling Huang
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Lu Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yunzi Chen
- Key Laboratory of Immune Microenvironment and Disease, Department of Immunology, Nanjing Medical University, Nanjing 211100, China.
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China.
| |
Collapse
|
7
|
Wang F, Gu X, Lin S, Wu Q, Sun Y, Zhang Q, Luo A, Feng X, Wang L, Xu L, Sun W, Tan W. Peptidase inhibitor 16 promotes inflammatory arthritis by suppressing Foxp3 expression via regulating K48-linked ubiquitin degradation Bmi-1 in regulatory T cells. Clin Immunol 2024; 259:109883. [PMID: 38147957 DOI: 10.1016/j.clim.2023.109883] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023]
Abstract
Abnormalities of regulatory T cells (Tregs) has been suggested in rheumatoid arthritis (RA), and Forkhead box P3 (Foxp3) is the key transcriptional factor of Tregs expression. However, the underlying molecular mechanism remains unclear. Here, we demonstrated peptidase inhibitor 16 (PI16) was significantly increased in the peripheral blood, synovial fluid, and synovial tissue from RA patients. PI16 transgenic mice (PI16Tg) aggravated arthritis severity partly through suppressing Foxp3 expression. Mechanistically, PI16 could interact with and stabilize Bmi-1 in Tregs via inhibiting K48-linked polyubiquitin of Bmi-1, which promotes the enrichment of repressive histone mark in Foxp3 promoter. Furthermore, Bmi-1 specific inhibitor PTC209 could restore Foxp3 expression and alleviate arthritis progression in PI16Tg mice, accompanied by increased recruitment of active histone mark in the promoter of Tregs. Our results suggest that PI16-Bmi-1 axis plays an important role in RA and other autoimmune diseases by suppressing Foxp3 expression in Tregs via Bmi-1-mediated histone modification.
Collapse
Affiliation(s)
- Fang Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xin Gu
- Department of Cardiology, the Affiliated Hospital of Jiangnan University, Wuxi 214125, China
| | - Shiyu Lin
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qin Wu
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuankai Sun
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qian Zhang
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Aishu Luo
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaoke Feng
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Integrated Traditional Chinese and Western Medicine Institute of Nanjing Medical University, Nanjing 210029, China
| | - Lei Wang
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lingxiao Xu
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Sun
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Wenfeng Tan
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
8
|
Wielińska J, Górna K, Świerkot J, Bugaj B, Kolossa K, Jeka S, Bogunia-Kubik K. Polymorphic Variants in the Vitamin D Receptor and Clinical Parameters of Rheumatoid Arthritis Patients Undergoing Anti-TNF Treatment. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0023. [PMID: 39522115 DOI: 10.2478/aite-2024-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Vitamin D levels have been related to the severity and progression of various autoimmune disorders. In this study, we aimed to investigate the impact of genetic variability in the vitamin D receptor (VDR) gene on disease susceptibility and progression in patients with rheumatoid arthritis (RA) treated with tumor necrosis factor (TNF) inhibitors. The study comprises 121 RA patients subjected to anti-TNF therapy genotyped for four VDR polymorphic variants: rs1544410 (BsmI), rs2228570 (FokI), rs731236 (TaqI), and rs7975232 (ApaI). There was no significant association between RA susceptibility and VDR genetic variants. The study results revealed that patients with the rs2228570 CC genotype were characterized by lower vitamin D3 levels (p = 0.028) than those with the T allele. Also, the vitamin D3 levels (p = 0.029) and age at diagnosis (p = 0.017) were significantly lower in rs7975232 A allele carriers compared to CC homozygotes. However, after 6 months of therapy, the A allele seemed to be related to lower disease activity score 28 (DAS28) values (p = 0.030) and more common in patients who achieved remission (p = 0.004) compared to the CC genotype. Concerning other investigated polymorphisms, patients carrying rs1544410 AA and rs731236 CC homozygosity had lower C-reactive protein (CRP) levels before therapy (p = 0.009). In conclusion, VDR rs2228570 and rs7975232 polymorphic variants were found to be related to vitamin D3 levels. Moreover, the genotyping of rs7975232 was also useful in evaluating disease onset and disease activity after 6 months of therapy with TNF inhibitors in RA patients.
Collapse
Affiliation(s)
- Joanna Wielińska
- Department of Clinical Immunology, Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland
| | - Katarzyna Górna
- Department of Clinical Immunology, Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland
| | - Jerzy Świerkot
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Bugaj
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Katarzyna Kolossa
- Department of Rheumatology and Connective Tissue Diseases, Jan Biziel University Hospital No. 2, Bydgoszcz, Poland
| | - Sławomir Jeka
- Department of Rheumatology and Connective Tissue Diseases, Jan Biziel University Hospital No. 2, Bydgoszcz, Poland
- Clinic and Department of Rheumatology and Connective Tissue Diseases, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Jagiellońska 15, 85-067 Bydgoszcz, Poland
| | - Katarzyna Bogunia-Kubik
- Department of Clinical Immunology, Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland
| |
Collapse
|
9
|
Lin X, Lin T, Wang X, He J, Gao X, Lyu S, Wang Q, Chen J. Sesamol serves as a p53 stabilizer to relieve rheumatoid arthritis progression and inhibits the growth of synovial organoids. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155109. [PMID: 37778247 DOI: 10.1016/j.phymed.2023.155109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/04/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease known as a leading cause of disability with considerable mortality. Developing alternative drugs and targets for RA treatment is an urgent issue. Sesamol is a phenolic compound isolated from natural food sesame (Sesamum indicum L.) with various biological activities. PURPOSE The current research intended to illuminate the bioactivity and mechanisms of sesamol in RA fibroblast-like synoviocytes (FLS), and aimed to estimate the potential clinical application value of sesamol in RA treatment. METHODS CCK-8, EdU, and flow cytometry assays, as well as transwell tests were applied to observe the effects of sesamol on the abnormal functions of RA-FLS. Moreover, synovial organoids and a collagen-induced arthritis (CIA) mouse model were constructed to further explore the therapeutic capacity of sesamol on RA. Furthermore, RNA sequencing combined with quantitative real-time PCR assay, Western blot as well as co-immunoprecipitation were employed to clarify the mechanism of sesamol in regulating RA progression. RESULTS Sesamol suppressed the proliferation through inhibiting DNA replication, triggering cell cycle arrest and apoptosis of RA-FLS. Besides, sesamol impaired RA-FLS migration and invasion. Interestingly, sesamol inhibited the growth of constructed synovial organoids and alleviated RA symptoms in CIA mice. Moreover, RNA sequencing further implicated p53 signaling as a downstream pathway of sesamol. Furthermore, sesamol was shown to decrease p53 ubiquitination and degradation, thereby activating p53 signaling. Finally, bioinformatics analyses also highlighted the importance of sesamol-regulated networks in the progression of RA. CONCLUSIONS Our investigation demonstrated that sesamol served as a novel p53 stabilizer to attenuate the abnormal functions of RA-FLS via facilitating the activation of p53 signaling. Moreover, our study highlighted that sesamol might be an effective lead compound or candidate drug and p53 could be a promising target for the therapy of RA.
Collapse
Affiliation(s)
- Xian Lin
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China
| | - Tengyu Lin
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China
| | - Xiaocheng Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China
| | - Jiaxin He
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China
| | - Xu Gao
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China
| | - Shuyan Lyu
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China.
| | - Jian Chen
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China; Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen 518036, China.
| |
Collapse
|
10
|
Liang L, Huang Y, Chen L, Shi Z, Wang H, Zhang T, Li Z, Mi J, Fan T, Lu Y, Chen F, Huang W, Hu K. Radioprotective efficacy of Astilbin in mitigating radiation-induced lung injury through inhibition of p53 acetylation. ENVIRONMENTAL TOXICOLOGY 2023; 38:2967-2980. [PMID: 37598414 DOI: 10.1002/tox.23931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
Radiation-induced lung injury (RILI) is a common side effect in thoracic tumor patients undergoing radiotherapy. At present, there is no ideal radio-protective agent which is widely used in RILI treatment. Astilbin (AST), a bioactive flavonoid, exhibits various biological effects, including anti-inflammatory, antioxidant, and anti-fibrotic activities, which partly result from reducing oxidative stress and inflammation in various pathogenic conditions. However, the protective efficacy of AST to ameliorate RILI has not been reported. In this study, we employed network pharmacology, RNA sequencing, and experimental evaluation to reveal the effects and pharmacological mechanism of AST to treat RILI in vivo and in vitro. We observed that AST reduced radiation-induced apoptosis, DNA damage, inflammatory reactions, and the reactive oxygen species (ROS) level in human normal lung epithelial cells BEAS-2B. Further study showed that AST treatment significantly ameliorated RILI by reducing the radiation-induced pathology changes and inflammatory reaction of lung tissue in C57BL/6J mice. Mechanistically, the expression of epithelial-mesenchymal transition (EMT) markers and radiation-triggered acetylation of the p53 protein were alleviated by AST treatment. Furthermore, AST alleviated the acetylation of p53 after intervention of Trichostatin A (TSA). Our data indicate that AST can alleviate RILI by inhibiting inflammatory reactions and the EMT process through decreasing the expression of p53 acetylation. In conclusion, our study suggests that AST has great potential to be a new protective and therapeutic compound for RILI.
Collapse
Affiliation(s)
- Lixing Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yaqin Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Liuyin Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Housheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhixun Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Jinglin Mi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Ting Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yushuang Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Fuli Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| |
Collapse
|
11
|
Wątroba M, Szewczyk G, Szukiewicz D. The Role of Sirtuin-1 (SIRT1) in the Physiology and Pathophysiology of the Human Placenta. Int J Mol Sci 2023; 24:16210. [PMID: 38003402 PMCID: PMC10671790 DOI: 10.3390/ijms242216210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Sirtuins, especially SIRT1, play a significant role in regulating inflammatory response, autophagy, and cell response to oxidative stress. Since their discovery, sirtuins have been regarded as anti-ageing and longevity-promoting enzymes. Sirtuin-regulated processes seem to participate in the most prevalent placental pathologies, such as pre-eclampsia. Furthermore, more and more research studies indicate that SIRT1 may prevent pre-eclampsia development or at least alleviate its manifestations. Having considered this, we reviewed recent studies on the role of sirtuins, especially SIRT1, in processes determining normal or abnormal development and functioning of the placenta.
Collapse
Affiliation(s)
| | | | - Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Medical University of Warsaw, Chałubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (G.S.)
| |
Collapse
|
12
|
Gu X, Zhao L, Ye J, Chen L, Sui C, Li B, Wang X, Zhang J, Du Y. 1,25(OH) 2D 3 ameliorates doxorubicin‑induced cardiomyopathy by inhibiting the NLRP3 inflammasome and oxidative stress. Exp Ther Med 2023; 26:413. [PMID: 37559932 PMCID: PMC10407981 DOI: 10.3892/etm.2023.12112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/16/2023] [Indexed: 08/11/2023] Open
Abstract
Doxorubicin (DOX), as a chemotherapy agent with marked therapeutic effect, can be used to treat certain types of cancer such as leukemia, lymphoma and breast cancer. However, the toxic effects of DOX on cardiomyocytes limit its clinical application. Oxidative stress has been documented to serve a pivotal role in DOX-induced cardiomyopathy. Previous studies have reported that 1,25(OH)2D3 has antioxidant and anti-inflammatory effects and can inhibit the renin-angiotensin system. However, the effects of 1,25(OH)2D3 on the pathophysiological processes of DOX-induced cardiomyopathy and its mechanisms remain poorly understood. To investigate these potential effects, C57BL/6J mice were used to construct a DOX-induced cardiomyopathy model and treated with 1,25(OH)2D3. At 4 weeks after the first injection of DOX, cardiac function and myocardial injury were evaluated by echocardiograph and ELISA. Masson's trichrome staining and RT-qPCR were used to assess myocardial fibrosis, and immunohistochemistry and western blotting were performed to analyze expression levels of inflammation and oxidative stress, and the NLRP3 inflammasome pathway. ChIP assay was used to assess the effects of 1,25(OH)2D3 on histone modification in the NLRP3 and Nrf2 promoters. The results showed that 1,25(OH)2D3 treatment increased LVEF and LVFS, reduced serum levels of BNP and cTnT, inhibited the collagen deposition and profibrotic molecular expression, and downregulated the levels of inflammatory cytokines in DOX-induced cardiomyopathy. ROS and antioxidant indices were also ameliorated after 1,25(OH)2D3 treatment. In addition, 1,25(OH)2D3 was found to inhibit the NLRP3 inflammasome and KEAP-Nrf2 pathways through regulation of the levels of H3K4me3, H3K27me3 and H2AK119Ub in the NLRP3 and Nrf2 promoters. In conclusion, the present study demonstrated that 1,25(OH)2D3 regulated histone modification in the NLRP3 and Nrf2 promoters, which in turn inhibits the activation of NLRP3 inflammasome and oxidative stress in cardiomyocytes, alleviating DOX-induced cardiomyopathy. Therefore, 1,25(OH)2D3 may be a potential drug candidate for the treatment of DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Lin Zhao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Jiabao Ye
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| | - Lin Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| | - Chenyan Sui
- Department of Neurology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Baihong Li
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Jun Zhang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| | - Yingqiang Du
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, P.R. China
| |
Collapse
|
13
|
Guo J, Ye W, Wu X, Huang H, Li B, Sun Z, Ren Z, Yang Z. TNF-α activates RELA expression via TNFRSF1B to upregulate OPA1 expression and inhibit chondrogenic differentiation of human adipose stem cells. J Orthop Surg Res 2023; 18:430. [PMID: 37312126 DOI: 10.1186/s13018-023-03846-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Tumor necrosis factor-alpha (TNF-α), one of the pro-inflammatory cytokines mediating the local inflammatory process in joints, inhibits cartilage formation and has a detrimental effect on stem cell-based cartilage regeneration for the treatment of osteoarthritis (OA). However, the mechanisms behind this inhibitory effect are still poorly understood. Mitochondrial morphological changes mediated by mitochondrial fusion and fission are highly plastic, are quite sensitive to environmental stimuli and play a crucial role in maintaining cell structure and function. In our study, chondrogenic differentiated human adipose stem cells (hADSCs) were exposed to TNF-α and the effect of TNF-α on the ability of hADSCs to chondrogenic differentiate and on mitochondrial fusion and fission was observed and analyzed. The aim was to investigate the role and mechanisms of mitochondrial fusion and fission regulation in the chondrogenic differentiation of hADSCs under normal conditions and under exposure to TNF-α. METHODS We used flow cytometry to identify hADSCs immunophenotypes CD29, CD44, CD34, CD45, and HLA-DR. Alcian blue staining and Sirius red staining were used to observe the formation of proteoglycans and collagen during the chondrogenic differentiation of hADSCs, respectively. The mRNA and protein expression levels of the cartilage formation marker SOX9, type II collagen (COL2A1), and Aggrecan were measured by real-time fluorescent quantitative PCR (RT-qPCR) and western blot, respectively. The fluorescent probes MitoTracker® Red CMXRos and JC-1 were used to visualize mitochondria morphology and detect mitochondrial membrane electricity (MMP). Affymetrix PrimeView™ chips were used for gene expression profiling. RESULTS The results showed that the chondrogenic differentiation of hADSCs was inhibited in the presence of TNF-α that optic atrophy 1 (OPA1) expression was significantly upregulated and mitochondria were prolonged and interconnected during this process. Gene microarray and RT-qPCR data showed that the presence of TNF-α led to increased expression of TNFα receptor 2 (TNFRSF1B) and RELA during chondrogenic differentiation of hADSCs. CONCLUSIONS TNF-α inhibits chondrogenic differentiation of human adipose stem cells by activating RELA expression through TNFRSF1B upregulating OPA1 expression thereby increasing mitochondrial fusion.
Collapse
Affiliation(s)
- Jiajia Guo
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Wang Ye
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xinglin Wu
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Haifeng Huang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Bo Li
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Zeyu Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Zhijing Ren
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| | - Zhen Yang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
14
|
Xu X, Luo H, Chen Q, Wang Z, Chen X, Li X, Chen H, Wang M, Xu Y, Dai M, Wang J, Huang X, Wu B, Li Y. Detecting potential mechanism of vitamin D in treating rheumatoid arthritis based on network pharmacology and molecular docking. Front Pharmacol 2022; 13:1047061. [DOI: 10.3389/fphar.2022.1047061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
Aim: Vitamin D plays a vital role in Rheumatoid arthritis (RA). However, the mechanism of vitamin D and rheumatism is still unclear. Therefore, a strategy based on network pharmacology and molecular docking was used to explore the mechanism of vitamin D and RA.Methods: The targets of RA were obtained from the GeneCards database and Therapeutic Targets Database, and the targets of vitamin D were obtained from the Drugbank database and STITCH database. Next, overlapping genes were identified by Venny, and further Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and molecular docking analyses were performed.Results: A total of 1,139 targets of RA and 201 targets of vitamin D were obtained. A total of 76 overlapping genes were identified by Venny. The enrichment analysis showed that cell proliferation, immune response, and apoptotic process were the critical biological processes of vitamin D in treating RA. Antifolate resistance, osteoclast differentiation, and the nuclear factor-kappa B (NF-κB) signalling pathway are fundamental mechanisms of vitamin D in treating RA. According to further molecular docking, ALB, TNF, CASP3, and TP53 may be important punctuation points or diagnostic markers for future RA treatment.Conclusion: By analysing overlapping genes of diseases and drugs, this study confirmed that ALB, TNF, CASP3, and TP53 may be essential markers or diagnostic markers for future RA treatment.
Collapse
|
15
|
Miao D, Goltzman D. Mechanisms of action of vitamin D in delaying aging and preventing disease by inhibiting oxidative stress. VITAMINS AND HORMONES 2022; 121:293-318. [PMID: 36707138 DOI: 10.1016/bs.vh.2022.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although several recent studies have shown that vitamin D supplementation beneficially decreases oxidative stress parameters, there is no consensus on this subject in humans. Thus the role of vitamin D supplementation has recently become a controversial topic because large intervention studies in humans have not shown significant benefits. These studies have indicated that supplementation with precursor forms of active vitamin D has no effect on all-cause mortality, cannot reduce the fracture risk of the elderly, cannot reduce the incidence of cancer or cardiovascular disease in the elderly, and cannot significantly reduce the incidence risk of diabetes in the elderly. However, a link between several age-related diseases and enhanced oxidative stress has been found in mice with insufficient or deficient 1,25-dihydroxyvitamin D (1,25(OH)2D), the active form of vitamin D, which indicates that reduced active vitamin D accelerates aging and age-related diseases by increasing oxidative stress. Furthermore, supplementation of exogenous 1,25(OH)2D3, or antioxidants, could dramatically postpone aging, prevent osteoporosis and spontaneous tumor development induced by 1,25(OH)2D insufficiency or deficiency, by inhibiting oxidative stress. Mechanistically, the antioxidative effects of 1,25(OH)2D3 are carried out via the vitamin D receptor (VDR) by activation of the Nrf2 oxidative stress response pathway though transcriptional or posttranscriptional activation of Nrf2 or transcriptional upregulation of Sirt1 and Bmi1 expression. Whether discrepancies between studies in humans and in mice reflect the different forms of vitamin D examined remains to be determined.
Collapse
Affiliation(s)
| | - David Goltzman
- McGill University Health Centre and McGill University, Montreal, QC, Canada.
| |
Collapse
|
16
|
Wang S, Du Q, Sun J, Geng S, Zhang Y. Investigation of the mechanism of Isobavachalcone in treating rheumatoid arthritis through a combination strategy of network pharmacology and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2022; 294:115342. [PMID: 35525528 DOI: 10.1016/j.jep.2022.115342] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 05/01/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Isobavachalcone (IBC) is a natural chalcone compound widely distributed in traditional Chinese medicine Psoralea corylifolia L., and Tibetan medicine Abelmoschus manihot (L.) Medik. Etc.. Among them, Psoralea corylifolia has the effect of tonifying the kidney and strengthening Yang, and it is recorded in the Medicinal theory that it can be used in managing rheumatism and arthralgia. In addition, It has been included in many prescriptions in traditional Chinese medicine as the main herb for managing rheumatoid arthritis (RA). Similarly, Abelmoschus manihot is a common Tibetan medicinal herb and is a common medicinal material in Tibetan medicine and reported in ancient medicinal books such as Jing Zhu Ben Cao and Si Bu Yi Dian to possess the effect of Ganhuangshui and thus can be used in treating Huangshui diseases (such as RA). Previous research has demonstrated IBC to possess numerous biological activities, including anti-cancer, anti-inflammatory, antibacterial and immunomodulatory. Nevertheless, its efficacy and potential mechanism in treating rheumatoid arthritis are yet to be investigated. AIM OF THE STUDY This study aimed at investigating the therapeutic efficacy and mechanism of IBC in treating RA through a combined strategy of network pharmacology, in vitro, and in vivo evaluation. MATERIALS AND METHODS The Swiss Target Prediction and GeneCards databases were consulted to predict the potential targets of IBC and RA. Additionally, the potential targets for IBC in treating RA were predicted by consulting databases such as String, Cytoscape, MCODE, and Cytohubba. R software was utilized for enrichment analysis of GO and KEGG pathways, followed by in vitro experimentation using cell lines and in vivo experimentation using animals to explore the potential mechanism of IBC in RA treatment. RESULTS By integrating the results of network pharmacological analysis, 17 genes were found to be strongly associated with RA, such as TNF, MAPK13, EGFR, PTGS2, MMP3, etc. The enrichment analysis indicated that IBC possessed tremendous therapeutic efficacy in managing RA through PI3K-AKT, rheumatoid arthritis, and TNF signaling pathways. The in vitro experimentation indicated that IBC inhibited the proliferation, migration, and invasion, and promoted apoptosis and inhibition of inflammation of MH7A cell lines stimulated with TNF-α. The IBC might also have an increasing effect on the intracellular ROS and reducing effect on the mitochondrial membrane potential. The western blotting results indicated that IBC markedly inhibited the expression of p-PI3K, p-AKT, p-JAK1, p-STAT3 and SOCS3 proteins in TNF-α stimulated MH7A cells. Furthermore, we found that IBC also significantly reduced paw swelling and arthritis severity in CIA model rats through in vivo animal studies. CONCLUSIONS In short, this study explored the effect of IBC by combining network pharmacology prediction with in vitro and in vivo experimentation. The results indicated that IBC exerts its anti-rheumatoid arthritis effect by regulating cell proliferation and survival via PI3K/AKT and JAK/STAT signaling pathways. This may open a new horizon and provide a theoretical foundation for further development and utilization of IBC in RA management.
Collapse
Affiliation(s)
- Shaohui Wang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qinyun Du
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sang Geng
- University of Tibetan Medicine, Lasa, 850000, China.
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
17
|
Vitamin D Supplementation in Patients with Juvenile Idiopathic Arthritis. Nutrients 2022; 14:nu14081538. [PMID: 35458099 PMCID: PMC9029755 DOI: 10.3390/nu14081538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 02/06/2023] Open
Abstract
Vitamin D has been implicated in the pathogenesis of skeletal disorders and various autoimmune disorders. Vitamin D can be consumed from the diet or synthesized in the skin upon ultraviolet exposure and hydroxylation in the liver and kidneys. In its bioactive form, vitamin D exerts a potent immunomodulatory effect and is important for bone health. Juvenile idiopathic arthritis (JIA) is a collection of inflammatory joint diseases in children that share the manifestation of inflamed synovium, which can result in growth arrest, articular deformity, bone density loss, and disability. To evaluate the potential effect of vitamin D on JIA disease manifestations and outcomes, we review the role of vitamin D in bone metabolism, discuss the mechanism of vitamin D in modulating the innate and adaptive immune systems, evaluate the clinical significance of vitamin D in patients with JIA, and summarize the supplementation studies.
Collapse
|
18
|
Wei K, Jiang P, Zhao J, Jin Y, Zhang R, Chang C, Xu L, Xu L, Shi Y, Guo S, He D. Biomarkers to Predict DMARDs Efficacy and Adverse Effect in Rheumatoid Arthritis. Front Immunol 2022; 13:865267. [PMID: 35418971 PMCID: PMC8995470 DOI: 10.3389/fimmu.2022.865267] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/07/2022] [Indexed: 12/28/2022] Open
Abstract
Rheumatoid arthritis (RA), one of the most common immune system diseases, mainly affects middle-aged and elderly individuals and has a serious impact on the quality of life of patients. Pain and disability caused by RA are significant symptoms negatively affecting patients, and they are especially seen when inappropriate treatment is administered. Effective therapeutic strategies have evolved over the past few decades, with many new disease-modifying antirheumatic drugs (DMARDs) being used in the clinic. Owing to the breakthrough in the treatment of RA, the symptoms of patients who could not be treated effectively in the past few years have been relieved. However, some patients complain about symptoms that have not been reported, implying that there are still some limitations in the RA treatment and evaluation system. In recent years, biomarkers, an effective means of diagnosing and evaluating the condition of patients with RA, have gradually been used in clinical practice to evaluate the therapeutic effect of RA, which is constantly being improved for accurate application of treatment in patients with RA. In this article, we summarize a series of biomarkers that may be helpful in evaluating the therapeutic effect and improving the efficiency of clinical treatment for RA. These efforts may also encourage researchers to devote more time and resources to the study and application of biomarkers, resulting in a new evaluation system that will reduce the inappropriate use of DMARDs, as well as patients' physical pain and financial burden.
Collapse
Affiliation(s)
- Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Yehua Jin
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Runrun Zhang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
- The Second Affiliated Hospital of the Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Linshuai Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Yiming Shi
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Shicheng Guo
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
19
|
Mitra S, Paul S, Roy S, Sutradhar H, Bin Emran T, Nainu F, Khandaker MU, Almalki M, Wilairatana P, Mubarak MS. Exploring the Immune-Boosting Functions of Vitamins and Minerals as Nutritional Food Bioactive Compounds: A Comprehensive Review. Molecules 2022; 27:555. [PMID: 35056870 PMCID: PMC8779769 DOI: 10.3390/molecules27020555] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 02/06/2023] Open
Abstract
Food components have long been recognized to play a fundamental role in the growth and development of the human body, conferring protective functionalities against foreign matter that can be severe public health problems. Micronutrients such as vitamins and minerals are essential to the human body, and individuals must meet their daily requirements through dietary sources. Micronutrients act as immunomodulators and protect the host immune response, thus preventing immune evasion by pathogenic organisms. Several experimental investigations have been undertaken to appraise the immunomodulatory functions of vitamins and minerals. Based on these experimental findings, this review describes the immune-boosting functionalities of micronutrients and the mechanisms of action through which these functions are mediated. Deficiencies of vitamins and minerals in plasma concentrations can lead to a reduction in the performance of the immune system functioning, representing a key contributor to unfavorable immunological states. This review provides a descriptive overview of the characteristics of the immune system and the utilization of micronutrients (vitamins and minerals) in preventative strategies designed to reduce morbidity and mortality among patients suffering from immune invasions or autoimmune disorders.
Collapse
Affiliation(s)
- Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (S.P.); (S.R.); (H.S.)
| | - Shyamjit Paul
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (S.P.); (S.R.); (H.S.)
| | - Sumon Roy
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (S.P.); (S.R.); (H.S.)
| | - Hriday Sutradhar
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (S.P.); (S.R.); (H.S.)
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh;
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Sulawesi Selatan, Indonesia;
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Selangor, Malaysia;
| | - Mohammed Almalki
- Department of Nursing, Faculty of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia;
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | | |
Collapse
|
20
|
Dai Y, Sheng J, He S, Wu Q, Wang Y, Su L. Dehydroevodiamine suppresses inflammatory responses in adjuvant-induced arthritis rats and human fibroblast-like synoviocytes. Bioengineered 2022; 13:268-279. [PMID: 34719315 PMCID: PMC8805850 DOI: 10.1080/21655979.2021.1999554] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/23/2021] [Indexed: 12/30/2022] Open
Abstract
Dehydroevodiamine (DHE) is an effective natural active substance extracted from Euodiae Fructus, which is a widely used herbal drug in traditional Chinese medicine. The focus of this study was to test the possibility of using DHE in the treatment of rheumatoid arthritis (RA) diseases. A rat model of adjuvant-induced arthritis (AIA) was generated using Complete Freund's Adjuvant (CFA). Body weight changes, arthritis scores, ankle pathology, tumor necrosis factor-alpha (TNF-α), interleukin-1β(IL-1β), interleukin-6 (IL-6), and interleukin-17 (IL-17) secretion, as well as matrix metalloproteinase (MMP) expression in joint tissue, were measured as indicators of viability of DHE medicated AIA rats. Human fibroblast-like synoviocytes (MH7A cells) were connected to check these impacts. The results confirmed that DHE administration had an excellent therapeutic impact on the AIA rat model, substantially relieving joint swelling, inhibiting synovial pannus hyperplasia, and decreasing joint scores. In addition, the serum enzyme-linked immunosorbent assay (ELISA) showed that DHE treatment reduced the expression of pro-inflammatory factors in AIA rats. The immunohistochemical results showed that DHE treatment could reduce the synthesis of MMPs such as matrix metalloproteinase-1(MMP-1) and matrix metalloproteinase-3 (MMP-3) in the ankle tissue of AIA rats. In vitro, DHE inhibited cell proliferation, mRNA transcription, protein synthesis of proinflammatory factors such as IL-1βand IL-6, and matrix metalloproteinases such as MMP-1 and MMP-3. Furthermore, DHE inhibited the phosphorylation levels of p38, JNK, and ERK proteins in TNF-α-treated MH7A cells.This work assessed the effect of DHE in AIA rats and revealed its mechanism in vitro.
Collapse
Affiliation(s)
- Yufang Dai
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Enshi, China
| | - Jiaoe Sheng
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Enshi, China
| | - Sanshan He
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Enshi, China
| | - Qingchao Wu
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Enshi, China
| | - Yunlong Wang
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Enshi, China
| | - Linchong Su
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Enshi, China
| |
Collapse
|
21
|
Romão VC, Fonseca JE. Etiology and Risk Factors for Rheumatoid Arthritis: A State-of-the-Art Review. Front Med (Lausanne) 2021; 8:689698. [PMID: 34901047 PMCID: PMC8661097 DOI: 10.3389/fmed.2021.689698] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is the most common systemic inflammatory rheumatic disease. It is associated with significant burden at the patient and societal level. Extensive efforts have been devoted to identifying a potential cause for the development of RA. Epidemiological studies have thoroughly investigated the association of several factors with the risk and course of RA. Although a precise etiology remains elusive, the current understanding is that RA is a multifactorial disease, wherein complex interactions between host and environmental factors determine the overall risk of disease susceptibility, persistence and severity. Risk factors related to the host that have been associated with RA development may be divided into genetic; epigenetic; hormonal, reproductive and neuroendocrine; and comorbid host factors. In turn, environmental risk factors include smoking and other airborne exposures; microbiota and infectious agents; diet; and socioeconomic factors. In the present narrative review, aimed at clinicians and researchers in the field of RA, we provide a state-of-the-art overview of the current knowledge on this topic, focusing on recent progresses that have improved our comprehension of disease risk and development.
Collapse
Affiliation(s)
- Vasco C Romão
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Centre and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal.,Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Centre and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal.,Rheumatology Research Unit, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
Zhou J, Hou C, Chen H, Qin Z, Miao Z, Zhao J, Wang Q, Cui M, Xie C, Wang R, Li Q, Zuo G, Miao D, Jin J. P16 I NK 4a Deletion Ameliorates Damage of Intestinal Epithelial Barrier and Microbial Dysbiosis in a Stress-Induced Premature Senescence Model of Bmi-1 Deficiency. Front Cell Dev Biol 2021; 9:671564. [PMID: 34712655 PMCID: PMC8545785 DOI: 10.3389/fcell.2021.671564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
This study aimed to determine whether Bmi-1 deficiency leads to intestinal epithelial barrier destruction and microbiota dysfunction, which members of the microbial community alter barrier function with age, and whether p16INK4a deletion could reverse the damage of intestinal epithelial barrier and microbial dysbiosis. Intestines from Bmi-1–deficient (Bmi-1–/–), Bmi-1 and p16INK4a double-knockout (Bmi-1–/–p16INK4a–/–), and wild-type mice were observed for aging and inflammation. Duolink Proximity Ligation Assay, immunoprecipitation, and construction of p16INK4a overexpressed adenovirus and the overexpressed plasmids of full-length, mutant, or truncated fragments for occludin were used for analyzing the interaction between p16INK4a and occludin. High-throughput sequencing of V4 region amplicon of 16S ribosomal RNA was conducted using intestinal microbiota. We found Bmi-1 deficiency destructed barrier structure, barrier function, and tight junction (TJ) in intestinal epithelium; decreased the TJ proteins; increased tumor necrosis factor α (TNF-α)–dependent barrier permeability; and up-regulated proinflammatory level of macrophages induced by intestinal microbial dysbiosis. The transplantation of fecal microbiota from wild-type mice ameliorated TJ in intestinal epithelium of Bmi-1–/– and Bmi-1–/–p16INK4a–/– mice. Harmful bacteria including Desulfovibrio, Helicobacter, and Oscillibacter were at a higher level in Bmi-1–/– mice. More harmful bacteria Desulfovibrio entered the epithelium and promoted macrophages-secreted TNF-α and caused TNF-α–dependent barrier permeability and aging. Accumulated p16INK4a combined with occludin at the 1st–160th residue in cytoplasm of intestinal epithelium cells from Bmi-1–/– mice, which blocked formation of TJ and the repair of intestinal epithelium barrier. P16INK4a deletion could maintain barrier function and microbiota balance in Bmi-1–/– mice through strengthening formation of TJ and decreasing macrophages-secreted TNF-α induced by Desulfovibrio entering the intestinal epithelium. Thus, Bmi-1 maintained intestinal TJ, epithelial barrier function, and microbiota balance through preventing senescence characterized by p16INK4a accumulation. The clearance of p16INK4a-positive cells in aging intestinal epithelium would be a new method for maintaining barrier function and microbiota balance. The residues 1–160 of occludin could be a novel therapeutic target for identifying small molecular antagonistic peptides to prevent the combination of p16INK4a with occludin for protecting TJ.
Collapse
Affiliation(s)
- Jiawen Zhou
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chenxing Hou
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Haiyun Chen
- Anti-Aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Ziyue Qin
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Zi'an Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jingyu Zhao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qiuyi Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Min Cui
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Rong Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, China
| | - Guoping Zuo
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Dengshun Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Anti-Aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, China
| | - Jianliang Jin
- Research Center for Bone and Stem Cells, Department of Human Anatomy, Key Laboratory for Aging and Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Zou J, Thornton C, Chambers ES, Rosser EC, Ciurtin C. Exploring the Evidence for an Immunomodulatory Role of Vitamin D in Juvenile and Adult Rheumatic Disease. Front Immunol 2021; 11:616483. [PMID: 33679704 PMCID: PMC7930375 DOI: 10.3389/fimmu.2020.616483] [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: 10/12/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Vitamin D is synthesized in the skin following exposure to UVB radiation or is directly absorbed from the diet. Following hydroxylation in the liver and kidneys, vitamin D becomes its bioactive form, 1,25(OH)2D, which has been described to have potent immunomodulatory capacity. This review will focus on the effect of vitamin D in modulating the dysregulated immune system of autoimmune rheumatic diseases (ARD) patients across age, in particular in arthritis (rheumatoid arthritis and juvenile idiopathic arthritis), and systemic lupus erythematosus (with adult and juvenile onset). As well as delineating the impact of vitamin D on the innate and adaptive immune functions associated with each disease pathology, this review will also summarize and evaluate studies that link vitamin D status with disease prevalence, and supplementation studies that examine the potential benefits of vitamin D on disease outcomes. Exploring this evidence reveals that better designed randomized controlled studies are required to clarify the impact of vitamin D supplementation on ARD outcomes and general health. Considering the accessibility and affordability of vitamin D as a therapeutic option, there is a major unmet need for evidence-based treatment recommendations for the use of vitamin D in this patient population.
Collapse
Affiliation(s)
- Jiaqi Zou
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - Clare Thornton
- Department of Rheumatology (Metabolic Bone Diseases), University College London Hospital NHS Foundation Trust, London, United Kingdom
| | - Emma S Chambers
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Elizabeth C Rosser
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London and Great Ormond Street Hospitals, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London and Great Ormond Street Hospitals, London, United Kingdom
| |
Collapse
|
24
|
Li S, Du J, Gan H, Chen J, Zhou Y, Tian J, Ling G, Li F. Resveratrol promotes apoptosis and G2/M cell cycle arrest of fibroblast-like synoviocytes in rheumatoid arthritis through regulation of autophagy and the serine-threonine kinase-p53 axis. Arch Med Sci 2021; 20:280-288. [PMID: 38414451 PMCID: PMC10895956 DOI: 10.5114/aoms/119022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/15/2020] [Indexed: 02/29/2024] Open
Abstract
Introduction Resveratrol, a polyphenol extracted from many plant species, has emerged as a promising pro-apoptotic agent in various cancer cells. However, the role of resveratrol in cell proliferation and apoptosis of fibroblast-like synoviocytes in rheumatoid arthritis (RA-FLS) is not fully understood. The study was aimed at elucidating the role of resveratrol in cell proliferation and apoptosis of RA-FLS and the underlying molecular mechanism. Material and methods Cultured RA-FLSs were subjected to tumour necrosis factor α (TNF-α). The cell proliferation was measured by Cell Counting Kit-8 assay. Cell apoptosis and cell cycle of RA-FLSs were determined by flow cytometry. The levels of apoptosis or autophagy or cell cycle-related protein were detected by immunoblot analysis. Results In our study, we confirmed that resveratrol reversed TNF-α mediated cell proliferation in RA-FLS. Meanwhile, resveratrol blocked cells at the G2/M stage and reduced the ratio of S phase cells through upregulation of p53 and consequently led to apoptotic cell death. Quite interestingly, we found that resveratrol reversed TNF-α-induced autophagy. Inhibition of autophagy by resveratrol or autophagy inhibitor or Beclin-1 siRNA suppressed TNF-α mediated cell survival and promoted cell apoptosis. However, the autophagy inducer rapamycin (RAPA) reversed the effect of resveratrol on autophagy and cell proliferation. Mechanistic studies revealed that resveratrol inhibited the activation of the phosphoinositide 3-kinases/serine-threonine kinase (PI3K/AKT) pathway. Inhibition of PI3K/AKT pathway by inhibitor LY294002 or resveratrol increased the expression of p53 and decreased the expression of cycle protein (cyclin B1), which further led to block cells in the G2/M arrest. Conclusions Our preliminary study indicated that resveratrol may suppress RA-FLS cell survival and promote apoptosis at least partly through regulation of autophagy and the AKT-p53 axis.
Collapse
Affiliation(s)
- Shu Li
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinfeng Du
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haina Gan
- Department of Rheumatology and Immunology, The First People's Hospital of Changde City, Changde, Hunan, China
| | - Jinwei Chen
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Zhou
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Tian
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guanghui Ling
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fen Li
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
25
|
L Bishop E, Ismailova A, Dimeloe S, Hewison M, White JH. Vitamin D and Immune Regulation: Antibacterial, Antiviral, Anti-Inflammatory. JBMR Plus 2021; 5:e10405. [PMID: 32904944 PMCID: PMC7461279 DOI: 10.1002/jbm4.10405] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Regulation of immune function continues to be one of the most well-recognized extraskeletal actions of vitamin D. This stemmed initially from the discovery that antigen presenting cells such as macrophages could actively metabolize precursor 25-hydroxyvitamin D (25D) to active 1,25-dihydroxyvitamin D (1,25D). Parallel observation that activated cells from the immune system expressed the intracellular vitamin D receptor (VDR) for 1,25D suggested a potential role for vitamin D as a localized endogenous modulator of immune function. Subsequent studies have expanded our understanding of how vitamin D exerts effects on both the innate and adaptive arms of the immune system. At an innate level, intracrine synthesis of 1,25D by macrophages and dendritic cells stimulates expression of antimicrobial proteins such as cathelicidin, as well as lowering intracellular iron concentrations via suppression of hepcidin. By potently enhancing autophagy, 1,25D may also play an important role in combatting intracellular pathogens such as M. tuberculosis and viral infections. Local synthesis of 1,25D by macrophages and dendritic cells also appears to play a pivotal role in mediating T-cell responses to vitamin D, leading to suppression of inflammatory T helper (Th)1 and Th17 cells, and concomitant induction of immunotolerogenic T-regulatory responses. The aim of this review is to provide an update on our current understanding of these prominent immune actions of vitamin D, as well as highlighting new, less well-recognized immune effects of vitamin D. The review also aims to place this mechanistic basis for the link between vitamin D and immunity with studies in vivo that have explored a role for vitamin D supplementation as a strategy for improved immune health. This has gained prominence in recent months with the global coronavirus disease 2019 health crisis and highlights important new objectives for future studies of vitamin D and immune function. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Emma L Bishop
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Aiten Ismailova
- Department of PhysiologyMcGill UniversityMontrealQuebecCanada
| | - Sarah Dimeloe
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
- Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
| | - Martin Hewison
- Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
| | - John H White
- Department of PhysiologyMcGill UniversityMontrealQuebecCanada
- Department of MedicineMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
26
|
Zhao XN, Bai ZZ, Li CH, Sheng CL, Li HY. The NK-1R Antagonist Aprepitant Prevents LPS-Induced Oxidative Stress and Inflammation in RAW264.7 Macrophages. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1943-1952. [PMID: 32546961 PMCID: PMC7246327 DOI: 10.2147/dddt.s244099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Background The macrophage is one of the most important types of immune cells that protect against harmful stimuli. Macrophage activation plays a pivotal role in the progression and development of various inflammatory diseases. The neurokinin 1 receptor (NK-1R) is a G protein-coupled receptor that plays an important role in inflammatory diseases. Aprepitant is a kind of NK-1R antagonist. The purpose of this study is to determine the protective effect of aprepitant in lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. Methods We examined the anti-inflammatory and anti-oxidant effects of aprepitant in LPS-treated RAW264.7 macrophages by using real-time PCR, ELISA, and Western blot analysis. We also assessed cellular oxidative stress signaling by measuring the levels of cellular MDA, total ROS, and NADPH oxidase expression. Cellular NO production was measured by DAF-FM DA staining. The inhibitory effect of aprepitant against NF-κB signaling was evaluated by luciferase assay and Western blot analysis. Results The expression of NK-1R is increased in LPS-induced macrophages, suggesting a potential role of the receptor in the inflammatory response. We show that aprepitant protects macrophages against oxidative stress by reducing the generation of ROS and the expression of NOX-4. Furthermore, aprepitant inhibits the secretion of pro-inflammatory cytokines and chemotactic factors by mediating the NF-κB signaling pathway. Conclusion The NK-1R receptor antagonist aprepitant acts as an anti-inflammatory agent, indicating that the blockage of the NK-1R pathway in macrophages has the potential to suppress inflammation.
Collapse
Affiliation(s)
- Xiao-Nan Zhao
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Zhen-Zi Bai
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Cheng-Hua Li
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Chuan-Lun Sheng
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Hong-Yan Li
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| |
Collapse
|
27
|
Rui X, Yang Y, Chen Q, Wu J, Chen J, Zhang Q, Ren R, Yin D. Imperative and effective reversion of synovial hyperplasia and cartilage destruction in rheumatoid arthritis through multiple synergistic effects of O 2 and Ca 2. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111058. [PMID: 32993999 DOI: 10.1016/j.msec.2020.111058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/24/2020] [Accepted: 05/04/2020] [Indexed: 01/06/2023]
Abstract
Abnormal synovial hyperplasia and cartilage destruction in a joint cavity are the key causes affecting the pain and disability in rheumatoid arthritis (RA) and, unfortunately, there exists no effective treatment for them. This investigation reports an effective reversion of the above pathological characteristics in RA owing to the use of a prolonged O2/Ca2+-supporting phototherapy hydrogel. The performed in vitro and in vivo experiments exhibit that the prolonged O2-supporting not only promotes the direct cell-killing effects of singlet oxygen, but also persistently blocks the pathological feedback between the abnormal proliferation of fibroblast-like synoviocyte and the local oxygen depletion. Furthermore, the Ca2+, which is the other decomposition product of the O2 donor, induces mitochondrial Ca2+ overload and endoplasmic reticulum Ca2+ disorder and triggers Ca2+-associated apoptosis and immunogenic cell death. In addition to these multiple synergistic effects on synovial hyperplasia, the prolonged Ca2+ support can also induce the regeneration of cartilage in RA affected joints. The present study may thus provide an effective therapeutic strategy for the prevention and reversion of joint lesions and the accompanying arthralgia and deformity in RA.
Collapse
Affiliation(s)
- Xue Rui
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, PR China; Engineering Technology Research Center of Modernized Pharmaceutics, Hefei, Anhui Province 230012, PR China; Key Laboratory of Xin' an Medicine, Ministry of Education, Hefei 230012, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Qingqing Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China
| | - Jingjing Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China
| | - Jing Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China
| | - Qingqing Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China
| | - Rongrong Ren
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, PR China; Engineering Technology Research Center of Modernized Pharmaceutics, Hefei, Anhui Province 230012, PR China; Key Laboratory of Xin' an Medicine, Ministry of Education, Hefei 230012, PR China.
| |
Collapse
|
28
|
Zhao M, He H, Yin J. CARD6 protects against collagen-induced rheumatoid arthritis in mice through attenuating the inflammatory response and joint destruction via suppression of TNFR1/TRAF2 signaling. Biochem Biophys Res Commun 2020; 526:1092-1099. [PMID: 32317187 DOI: 10.1016/j.bbrc.2020.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/03/2020] [Indexed: 10/24/2022]
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases, characterized by chronic inflammation and bone destruction. However, the pathogenesis that contributes to RA is still unclear. Caspase recruitment domain protein 6 (CARD6) is a typical member of CARD domain-containing proteins, and shows regulatory effects on nuclear factor-κB (NF-κB) activation to meditate inflammation. In the present study, the role of CARD6 in the progression of inflammatory bone erosion in RA was investigated using the in vitro and in vivo experiments. In vitro results indicated that CARD expression was markedly down-regulated in the activated macrophages induced by lipopolysaccharide (LPS), accompanied with time-dependently increased expression of pro-inflammatory cytokines. Notably, over-expressing CARD6 in macrophages by adenoviral (Ad) vector significantly abolished the expression levels of pro-inflammatory cytokines and chemokines. We found that CARD6 over-expression-suppressed inflammatory response was associated with the blockage of tumor necrosis factor receptor-1/tumor necrosis factor receptor-associated factor-2 (TNFR1/TRAF2) signaling, inhibiting NF-κB pathway subsequently. In addition, LPS-induced apoptosis in macrophages was also blunted due to AdCARD6 infection. CARD6-alleviated inflammatory response and apoptotic cell death were further confirmed in TNF-α-stimulated macrophages. Then, the in vivo studies showed that promoting CARD6 expression using adeno-associated virus (AAV) effectively attenuated the severity of arthritis, improved histopathological damage, and hindered the bone erosion in collagen-induced arthritis (CIA) mice. Moreover, pro-inflammatory factors in the joint samples were also markedly decreased in CIA mice with CARD6 over-expression, which was related to the down-regulation of TNFR1/TRAF2/NF-κB signaling pathway. Meanwhile, apoptosis in joint of CIA mice was also ameliorated by AAV-CARD6, as evidenced by the obviously reduced expression of cleaved Caspase-3. These results clearly suggested that CARD6 might have anti-inflammatory and anti-apoptotic effects during RA progression, and thus could be defined as a novel therapeutic target for RA treatment in future.
Collapse
Affiliation(s)
- Min Zhao
- Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, 471002, China
| | - Hongying He
- Department of Orthopedics, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Jianbao Yin
- Department of Rheumatology, Affiliated Tengzhou Central People's Hospital of Jining Medical University, Jining Medical University, Tengzhou City, Shandong Province, Tengzhou, 277500, China.
| |
Collapse
|
29
|
Bellan M, Andreoli L, Mele C, Sainaghi PP, Rigamonti C, Piantoni S, De Benedittis C, Aimaretti G, Pirisi M, Marzullo P. Pathophysiological Role and Therapeutic Implications of Vitamin D in Autoimmunity: Focus on Chronic Autoimmune Diseases. Nutrients 2020; 12:E789. [PMID: 32192175 PMCID: PMC7146294 DOI: 10.3390/nu12030789] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
Vitamin D is a pleiotropic secosteroid yielding multiple actions in human physiology. Besides the canonical regulatory activity on bone metabolism, several non-classical actions have been described and the ability of vitamin D to partake in the regulation of the immune system is particularly interesting, though far stronger and convincing evidence has been collected in in vitro as compared to in vivo studies. Whether vitamin D is able to regulate at physiological concentrations the human immune system remains unproven to date. Consequently, it is not established if vitamin D status is a factor involved in the pathogenesis of immune-mediated diseases and if cholecalciferol supplementation acts as an adjuvant for autoimmune diseases. The development of autoimmunity is a heterogeneous process, which may involve different organs and systems with a wide range of clinical implications. In the present paper, we reviewed the current evidences regarding vitamin D role in the pathogenesis and management of different autoimmune diseases.
Collapse
Affiliation(s)
- Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Laura Andreoli
- Rheumatology and Clinical Immunology Unit and Department of Clinical and Experimental Sciences, Spedali Civili and University of Brescia, 25128 Brescia, Italy; (L.A.); (S.P.)
| | - Chiara Mele
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Cristina Rigamonti
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical Immunology Unit and Department of Clinical and Experimental Sciences, Spedali Civili and University of Brescia, 25128 Brescia, Italy; (L.A.); (S.P.)
| | - Carla De Benedittis
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Gianluca Aimaretti
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of Internal Medicine, “AOU Maggiore della Carità”, 28100 Novara, Italy
- CAAD, Centre for Autoimmune and Allergic Diseases, 28100 Novara, Italy
| | - Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale UPO, 28100 Novara, Italy; (M.B.); (C.M.); (P.P.S.); (C.R.); (C.D.B.); (G.A.); (M.P.)
- Division of General Medicine, Ospedale S. Giuseppe, I.R.C.C.S. Istituto Auxologico Italiano, 28921 Verbania, Italy
| |
Collapse
|
30
|
Ding L, Sui X, Yang M, Zhang Q, Sun S, Zhu F, Cheng H, Zhang C, Chen H, Ding R, Cao J. Toxicity of cooking oil fume derived particulate matter: Vitamin D 3 protects tubule formation activation in human umbilical vein endothelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 188:109905. [PMID: 31706245 DOI: 10.1016/j.ecoenv.2019.109905] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 06/10/2023]
Abstract
Cooking oil fumes-derived PM2.5 (COFs-derived PM2.5) is the main source of indoor pollution. Exposure to COFs-derived PM2.5 can cause oxidative stress and affect angiogenesis. Here we investigated the roles of vitamin D3 (VD3) in protecting tubule formation injury induced by COFs-derived PM2.5, and the roles of ROS/NLRP3/VEGF signaling pathway in the effects. Human umbilical vein endothelial cells (HUVECs) were exposed to 0 (1‰ DMSO), 1000 nmol/l VD3, 100 μg/ml PM2.5, and 1000 nmol/l VD3 + 100 μg/ml PM2.5, respectively. Cell viability and tube formation, as well as protein and mRNA levels were measured. The results showed that exposure of COFs-derived PM2.5 dose-and time-dependently reduced the viability of HUVECs, increased the levels of mitochondrial and intracellular ROS, and changed the mitochondrial membrane potential level. While co-incubation with VD3 rescued these adverse effects. Both Western blot and real-time PCR (RT-PCR) showed that the expressions of NLRP3, caspase-1, Interleukin (IL)-1β, and IL-18 in COFs-derived PM2.5 exposure group increased significantly, which could be effectively decreased by co-incubation with VD3. COFs-derived PM2.5 exposure could also reduce the expression of VEGF, while co-incubating HUVECs with VD3 evidently up-regulated the protein level of VEGF in HUVECs. In addition, COFs-derived PM2.5 could also inhibit the tube formation of HUVECs in vitro, which could be effectively rescued by the co-incubation of VD3. Our study proved that COFs-derived PM2.5 could damage the tubule formation of HUVECs in vitro, which could be effectively rescue by co-incubation with VD3, in which processes the ROS/NLRP3/VEGF signaling pathway played a crucial role. It provides a new theoretical basis for further study on the toxicity of PM2.5 to umbilical cord blood vessels.
Collapse
Affiliation(s)
- Liu Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Xinmiao Sui
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Mei Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Qi Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Shu Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Furong Zhu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Han Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Chao Zhang
- Department of Environmental Health, Center for Disease Control and Prevention, Nanjing, China.
| | - Hongbo Chen
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical Universit, 15 Yimin Road, Hefei, China.
| | - Rui Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - Jiyu Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China; Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
31
|
Harrison SR, Li D, Jeffery LE, Raza K, Hewison M. Vitamin D, Autoimmune Disease and Rheumatoid Arthritis. Calcif Tissue Int 2020; 106:58-75. [PMID: 31286174 PMCID: PMC6960236 DOI: 10.1007/s00223-019-00577-2] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/18/2019] [Indexed: 02/06/2023]
Abstract
Vitamin D has been reported to influence physiological systems that extend far beyond its established functions in calcium and bone homeostasis. Prominent amongst these are the potent immunomodulatory effects of the active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3). The nuclear vitamin D receptor (VDR) for 1,25-(OH)2D3 is expressed by many cells within the immune system and resulting effects include modulation of T cell phenotype to suppress pro-inflammatory Th1 and Th17 CD4+ T cells and promote tolerogenic regulatory T cells. In addition, antigen-presenting cells have been shown to express the enzyme 1α-hydroxylase that converts precursor 25-hydroxyvitamin D3 (25-OHD3) to 1,25-(OH)2D3, so that immune microenvironments are able to both activate and respond to vitamin D. As a consequence of this local, intracrine, system, immune responses may vary according to the availability of 25-OHD3, and vitamin D deficiency has been linked to various autoimmune disorders including rheumatoid arthritis (RA). The aim of this review is to explore the immune activities of vitamin D that impact autoimmune disease, with specific reference to RA. As well as outlining the mechanisms linking vitamin D with autoimmune disease, the review will also describe the different studies that have linked vitamin D status to RA, and the current supplementation studies that have explored the potential benefits of vitamin D for prevention or treatment of RA. The overall aim of the review is to provide a fresh perspective on the potential role of vitamin D in RA pathogenesis and treatment.
Collapse
Affiliation(s)
- Stephanie R Harrison
- Institute of Metabolism and Systems Research, The University of Birmingham, Birmingham, B15 2TT, UK
- Department of Rheumatology, Sandwell and West, Birmingham Hospitals NHS Trust, Birmingham, B18 7QH, UK
| | - Danyang Li
- Institute of Metabolism and Systems Research, The University of Birmingham, Birmingham, B15 2TT, UK
| | - Louisa E Jeffery
- Institute of Translation Medicine, The University of Birmingham, Birmingham, B15 2TT, UK
| | - Karim Raza
- Department of Rheumatology, Sandwell and West, Birmingham Hospitals NHS Trust, Birmingham, B18 7QH, UK
- Institute of Inflammation and Ageing, Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence and MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Martin Hewison
- Institute of Metabolism and Systems Research, The University of Birmingham, Birmingham, B15 2TT, UK.
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TT, UK.
| |
Collapse
|
32
|
Chen H, Chen H, Liang J, Gu X, Zhou J, Xie C, Lv X, Wang R, Li Q, Mao Z, Sun H, Zuo G, Miao D, Jin J. TGF-β1/IL-11/MEK/ERK signaling mediates senescence-associated pulmonary fibrosis in a stress-induced premature senescence model of Bmi-1 deficiency. Exp Mol Med 2020; 52:130-151. [PMID: 31959867 PMCID: PMC7000795 DOI: 10.1038/s12276-019-0371-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 01/22/2023] Open
Abstract
To study whether TGF-β1/IL-11/MEK/ERK (TIME) signaling mediates senescence-associated pulmonary fibrosis (SAPF) in Bmi-1-deficient (Bmi-1-/-) mice and determines the major downstream mediator of Bmi-1 and crosstalk between p16INK4a and reactive oxygen species that regulates SAPF, phenotypes were compared among 7-week-old p16INK4a and Bmi-1 double-knockout, N-acetylcysteine (NAC)-treated Bmi-1-/-, Bmi-1-/-, and wild-type mice. Pulmonary fibroblasts and alveolar type II epithelial (AT2) cells were used for experiments. Human pulmonary tissues were tested for type Ι collagen, α-smooth muscle actin (α-SMA), p16INK4a, p53, p21, and TIME signaling by using enzyme-linked immunosorbent assay (ELISA). Our results demonstrated that Bmi-1 deficiency resulted in a shortened lifespan, ventilatory resistance, poor ventilatory compliance, and SAPF, including cell senescence, DNA damage, a senescence-associated secretory phenotype and collagen overdeposition that was mediated by the upregulation of TIME signaling. The signaling stimulated cell senescence, senescence-related secretion of TGF-β1 and IL-11 and production of collagen 1 by pulmonary fibroblasts and the epithelial-to-mesenchymal transition of AT2 cells. These processes were inhibited by anti-IL-11 or the MEK inhibitor PD98059. NAC treatment prolonged the lifespan and ameliorated pulmonary dysfunction and SAPF by downregulating TIME signaling more than p16INK4a deletion by inhibiting oxidative stress and DNA damage and promoting ubiquitin-proteasome degradation of p16INK4a and p53. Cytoplasmic p16INK4a accumulation upregulated MEK/ERK signaling by inhibiting the translocation of pERK1/2 (Thr202/Tyr204) from the cytoplasm to the nucleus in senescent fibroblasts. The accumulation of collagen 1 and α-SMA in human lungs accompanied by cell senescence may be mediated by TIME signaling. Thus, this signaling in aging fibroblasts or AT2 cells could be a therapeutic target for preventing SAPF.
Collapse
Affiliation(s)
- Haiyun Chen
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Anti-aging Research Laboratory, Friendship Plastic Surgery Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Hongjie Chen
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jialong Liang
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Gu
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jiawen Zhou
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xianhui Lv
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Rong Wang
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, Jiangsu, 210029, China
| | - Zhiyuan Mao
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Haijian Sun
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guoping Zuo
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dengshun Miao
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianliang Jin
- Research Center for Bone and Stem Cells, Department of Human Anatomy; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
33
|
Liu X, Zhu Y, Zheng W, Qian T, Wang H, Hou X. Antagonism of NK-1R using aprepitant suppresses inflammatory response in rheumatoid arthritis fibroblast-like synoviocytes. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1628-1634. [PMID: 31010320 DOI: 10.1080/21691401.2019.1573177] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Chronic inflammation in fibroblast-like synoviocytes (FLSs) induced by pro-inflammatory cytokines such as TNF-α plays a key role in the pathogenesis of rheumatoid arthritis (RA). The neurokinin-1 receptor (NK-1R) is one of the G protein-coupled receptors (GPCRs) mediating the intracellular signalling of substance P (SP). However, the possible implications of NK-1R in rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) and the pathogenesis of RA have not yet been reported. In the current study, we report that NK-1R is expressed in FLSs. Importantly, NK-1R expression was found to be significantly increased in RA-FLSs compared to normal FLSs. Interestingly, we found that treatment with tumour necrosis factor (TNF)-α increased the expression of NK-1R at both the gene and protein levels. Treatment with the NK-1R antagonist aprepitant reduced TNF-α-induced expression of NADPH oxidase 4 (NOX-4) and generation of reactive oxygen species (ROS) in FLSs. Our results also display that blockage of NF-1R using aprepitant inhibited TNF-α-induced expression and secretion of proinflammatory cytokines, including interleukin-1β (IL-1β), IL-6, and IL-8. Consistently, aprepitant prevented TNF-α-induced expression of matrix metalloproteinases (MMPs), including matrix metalloproteinase-3 (MMP-3) and matrix metalloproteinase-13 (MMP-13). Mechanistically, our data demonstrate that treatment with aprepitant inhibited TNF-α-induced phosphorylation and degradation of inhibitor of NF-κB (IκBα). Notably, aprepitant attenuated TNF-α-induced nuclear translocation of nuclear factor κB (NF-κB) p65 and reduced luciferase activity of NF-κB in FLSs. The findings implicated a novel function of NK-1R in RA-FLSs. Blockage of NK-1R using its specific antagonist aprepitant might provide a new therapeutic strategy for RA.
Collapse
Affiliation(s)
- Xiaoping Liu
- a Department of Rheumatology , Dongfang Hospital of Beijing University of Chinese Medicine , Beijing , China
| | - Yuelan Zhu
- a Department of Rheumatology , Dongfang Hospital of Beijing University of Chinese Medicine , Beijing , China
| | - Wei Zheng
- b Internal medicine of TCM , Beijing University of Chinese Medicine , Beijing , China
| | - Tangliang Qian
- b Internal medicine of TCM , Beijing University of Chinese Medicine , Beijing , China
| | - Haiyu Wang
- b Internal medicine of TCM , Beijing University of Chinese Medicine , Beijing , China
| | - Xiujuan Hou
- a Department of Rheumatology , Dongfang Hospital of Beijing University of Chinese Medicine , Beijing , China
| |
Collapse
|
34
|
Zhou W, Xu J, Wang C, Shi D, Yan Q. miR-23b-3p regulates apoptosis and autophagy via suppressing SIRT1 in lens epithelial cells. J Cell Biochem 2019; 120:19635-19646. [PMID: 31338869 DOI: 10.1002/jcb.29270] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/30/2019] [Indexed: 12/13/2022]
Abstract
Age-related cataract is one of the prior causes of blindness and the incidence rates of cataract are even rising. Oxidative stress plays an important role in the pathogenesis of cataracts. Under oxidative stress, lens epithelial cell (LEC cell) apoptosis is activated, which might lead to the opacity of the lens and accelerate the progression of cataract development. Meanwhile, autophagy is also active to face oxidative stress. miRNAs have been reported to involve cataract. However, the underlying mechanism is not clear. The present study aimed to investigate the regulatory effect of miR23b-3p on apoptosis and autophagy in LEC cells under oxidative stress. The expression levels of miR-23b-3p were examined in age-related cataract tissues and LEC cells treated with hydrogen peroxide, showing that miR23b-3p expression levels were upregulated. Knockdown of miR23b-3p expression in LEC cells brought about apoptosis significantly decreased while autophagy significantly increased during hydrogen peroxide. We predicted microRNA miRNA-23b-3p might participate in regulating silent information regulator 1 (SIRT1) by bioinformatics database of TargetScan. Luciferase reporter assays confirmed that miRNA-23b-p could suppress SIRT1 expression by binding its 3'UTR. In addition, overexpression or knockdown of miR-23b-3p could decrease or increase SIRT1 expression, which indicated that Mir-23b-3p could suppress SIRT1 expression. In addition, enhanced SIRT1 could attenuate the regulation of cell apoptosis and autophagy induced by overexpression of miR-23b-3p. Taken together, our findings revealed that miR-23b-3p regulated apoptosis and autophagy via suppressing SIRT1 in LEC cell under oxidative stress, which could provide new ideas for clinical treatment of cataract.
Collapse
Affiliation(s)
- Wenkai Zhou
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jun Xu
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Chunxia Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Dong Shi
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Qichang Yan
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
35
|
Liu J, Jiang T, Li C, Wu Y, He M, Zhao J, Zheng L, Zhang X. Bioconjugated Carbon Dots for Delivery of siTnfα to Enhance Chondrogenesis of Mesenchymal Stem Cells by Suppression of Inflammation. Stem Cells Transl Med 2019; 8:724-736. [PMID: 30919586 PMCID: PMC6591550 DOI: 10.1002/sctm.18-0289] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/20/2019] [Indexed: 01/05/2023] Open
Abstract
Although a promising strategy, the mesenchymal stem cell (MSC)-based therapy of cartilage defects is sometimes accompanied with chronic inflammation during the remodeling status, which may hinder cartilage regeneration. During this process, the inflammatory cytokine tumor necrosis factor α (TNFα) plays an important role and may be a potential target. In this study, we investigated the effect of Tnfα RNA interference by introducing a functional and highly safe carbon dot (CD)-SMCC nanovector synthesized by bioconjugation of CDs with a protein crosslinker, sulfosuccinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate (sulfo-SMCC), as the vehicle of the silenced TNFα (siTnfα) on chondrogenesis of MSCs. The results showed that CD-SMCC displayed intense fluorescence with well-dispersed and positively charged properties, which favored effective binding and delivering of siTnfα into the MSCs. CD-SMCC-siTnfα nanoformula also exhibited considerably high transfection efficiency and nearly no cytotoxicity, which is preferred over commercial polyethyleneimine. Interference of Tnfα by CD-SMCC-siTnfα markedly promoted the chondrogenesis of MSCs, as indicated by upregulating cartilage-specific markers. Furthermore, in vivo exploration indicated that CD-SMCC-siTnfα transfected MSCs accelerated cartilage regeneration. In conclusion, this study demonstrated that in combination with the novel CD-SMCC nanovector, targeting Tnfα may facilitate stem cell-based therapy of cartilage defects. Stem Cells Translational Medicine 2019;8:724&736.
Collapse
Affiliation(s)
- Jianwei Liu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
- Department of Spine OsteopathiaThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
- Department of Bone and Joint SurgeryThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Chun Li
- Department of Spine OsteopathiaThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Yang Wu
- Department of Orthopaedics Trauma and Hand SurgeryThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Maolin He
- Department of Spine OsteopathiaThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
- Department of Spine OsteopathiaThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
- Department of Bone and Joint SurgeryThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
- Department of Orthopaedics Trauma and Hand SurgeryThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for BiomedicineThe First Affiliated Hospital of Guangxi Medical University, Guangxi Medical UniversityNanningPeople's Republic of China
| | - Xingdong Zhang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduPeople's Republic of China
| |
Collapse
|
36
|
Vitamin D as a Principal Factor in Mediating Rheumatoid Arthritis-Derived Immune Response. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3494937. [PMID: 31205940 PMCID: PMC6530219 DOI: 10.1155/2019/3494937] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic multifactorial autoimmune disorder. The interactions between diverse environmental and genetic factors lead to the onset of this complex autoimmune disorder. Serum levels of vitamin D (VD) are involved in the regulation of various immune responses. Vitamin D is a key signaling molecule in the human body that maintains calcium as well as phosphate homeostasis. It also regulates the functions of the immune system and, thus, can play a substantial role in the etiology of various autoimmune disorders, including RA. Low serum VD levels have been found to be associated with a higher risk of RA, although this finding has not been replicated consistently. The molecular mechanisms by which VD influences autoimmunity need to be further explored to understand how variation in plasma VD levels could affect the pathogenesis of RA. This mini-review focuses on the influence of VD and its serum levels on RA susceptibility, RA-associated complexities, treatment, and transcriptome products of key proinflammatory cytokines, along with other cytokines that are key regulators of inflammation in rheumatoid joints.
Collapse
|
37
|
Yamamoto E, Jørgensen TN. Immunological effects of vitamin D and their relations to autoimmunity. J Autoimmun 2019; 100:7-16. [PMID: 30853311 DOI: 10.1016/j.jaut.2019.03.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/27/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Abstract
Vitamin D deficiency is an established risk factor for many autoimmune diseases and the anti-inflammatory properties of vitamin D underscore its potential therapeutic value for these diseases. However, results of vitamin D3 supplementation clinical trials have been varied. To understand the clinical heterogeneity, we reviewed the pre-clinical data on vitamin D activity in four common autoimmune diseases: multiple sclerosis (MS), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD), in which patients are commonly maintained on oral vitamin D3 supplementation. In contrast, many pre-clinical studies utilize other methods of manipulation (i.e. genetic, injection). Given the many actions of vitamin D3 and data supporting a vitamin D-independent role of the Vitamin D receptor (VDR), a more detailed mechanistic understanding of vitamin D3 activity is needed to properly translate pre-clinical findings into the clinic. Therefore, we assessed studies based on route of vitamin D3 administration, and identified where discrepancies in results exist and where more research is needed to establish the benefit of vitamin D supplementation.
Collapse
Affiliation(s)
- Erin Yamamoto
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Trine N Jørgensen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA.
| |
Collapse
|
38
|
Qian X, Zhao Z, Shang W, Xu Z, Zhang B, Cai H. Serum proteomic analysis of the anti‑arthritic effects of sinomenine on rats with collagen‑induced arthritis. Mol Med Rep 2018; 18:49-58. [PMID: 29749546 PMCID: PMC6059692 DOI: 10.3892/mmr.2018.8959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/16/2018] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease, which is characterized by inflammatory synovitis, and the subsequent destruction of articular cartilage and bone. Sinomenine is a traditional Chinese medicine, which has been employed as a clinical treatment for RA for several years in China. The present study investigated the anti‑arthritic effects of sinomenine on Sprague‑Dawley rats with collagen‑induced arthritis (CIA). The differentially expressed proteins in serum were measured by proteomic analysis in order to generate a differentially expressed protein network. A total of 320 differentially expressed proteins were detected in the drug‑treated group compared with in the control group. In the sinomenine‑treated group, 79 differentially expressed proteins were detected compared with in the model group, and among these, 46 proteins were upregulated. Gene ontology analysis revealed that five functions were affected by sinomenine treatment of CIA rats compared with in the model group. In addition, Ingenuity® Pathway Analysis was used to measure enriched signaling pathways, which revealed nuclear factor‑κB, histones, heat shock proteins and protein kinase B as core proteins, generating ~60 pair associations in the network. To the best of our knowledge, the present study is the first to perform proteomic analysis in sinomenine‑treated CIA rats, and the results revealed that numerous targets were involved in the process. In addition, the present study provided a novel approach and evidence for exploring the biological effects of sinomenine. Therefore, the findings of the present study may provide a novel insight into the anti‑RA mechanisms of sinomenine, and may justify further exploration into its function in other relevant diseases.
Collapse
Affiliation(s)
- Xin Qian
- Department of Nursing, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Zhiming Zhao
- Integrated Traditional Chinese and Western Medicine, Nanjing General Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Shang
- Integrated Traditional Chinese and Western Medicine, Nanjing General Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Zhihan Xu
- Department of Nursing, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Beibei Zhang
- Integrated Traditional Chinese and Western Medicine, Nanjing General Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Hui Cai
- Integrated Traditional Chinese and Western Medicine, Nanjing General Hospital, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
39
|
Lu B, Christensen IT, Ma LW, Wang XL, Jiang LF, Wang CX, Feng L, Zhang JS, Yan QC. miR-211 promotes lens epithelial cells apoptosis by targeting silent mating-type information regulation 2 homolog 1 in age-related cataracts. Int J Ophthalmol 2018; 11:201-207. [PMID: 29487807 DOI: 10.18240/ijo.2018.02.04] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/21/2017] [Indexed: 01/12/2023] Open
Abstract
AIM To detect the expression of miR-211 in age-related cataract tissue, explore the effects of miR-211 on lens epithelial cell proliferation and apoptosis, and identify its target gene. METHODS This study used real-time quantitative polymerase chain reaction (RT-qPCR) to measure the expression of miR-211 and its predicted target gene [silent mating-type information regulation 2 homolog 1 (SIRT1)] in 46 anterior lens capsules collected from age-related cataract patients. Human lens epithelial cell line (SRA01/04) cells were transfected with either miR-211 mimics, mimic controls, miR-211 inhibitors or inhibitor controls, 72h after transfection, miRNA and protein expression of SIRT1 were measured using RT-qPCR and Western blotting; then cells were exposed to 200 µmol/L H2O2 for 1h, whereupon cell viability was measured by MTS assay, caspase-3 assay was performed. Dual luciferase reporter assay was performed to verify the relationship between miR-211 of SIRT1. RESULTS Compared to the control group, expression of miR-211 was significantly increased (P<0.001), the miRNA and protein expression of SIRT1 were significantly decreased (P<0.001) in the anterior lens capsules of patients with age-related cataracts. Relative to the control group, SIRT1 miRNA and protein levels in the miR-211 mimic group were significantly reduced, cell proliferation activity significantly decreased, and caspase-3 activity was significantly increased (P<0.001). In the miR-211 inhibitor group, SIRT1 miRNA and protein expression were significantly increased, cell proliferation activity significantly increased, and caspase-3 activity was significantly decreased (P<0.001). A dual luciferase reporter assay confirmed that SIRT1 is a direct target of miR-211. CONCLUSION miR-211 is highly expressed in the anterior lens capsules of patients with age-related cataracts. By negatively regulating the expression of SIRT1, miR-211 promotes lens epithelial cell apoptosis and inhibits lens epithelial cell proliferation.
Collapse
Affiliation(s)
- Bo Lu
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Ian T Christensen
- University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | - Li-Wei Ma
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Xin-Ling Wang
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Ling-Feng Jiang
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Chun-Xia Wang
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Li Feng
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Jin-Song Zhang
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| | - Qi-Chang Yan
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Key Laboratory of Lens Research of Liaoning Province, Eye Hospital of China Medical University, Shenyang 110005, Liaoning Province, China
| |
Collapse
|
40
|
Zhu JJ, Wang ZQ, Bi LQ, Liu GF. Role of lentivirus-mediated overexpression of SOCS3 in proliferation and apoptosis of fibroblasts-like synoviocytes in Lewis rats with adjuvant-induced rheumatoid arthritis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:981-989. [PMID: 31938192 PMCID: PMC6958020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 12/16/2017] [Indexed: 06/10/2023]
Abstract
The purpose of this study is to explore the effects of lentivirus-mediated overexpression of the SOCS3 gene on proliferation and apoptosis of fibroblasts-like synoviocytes (FLSs) in rheumatoid arthritis (RA). A total of 20 Lewis rats were randomly assigned into experimental and normal groups. Rats in the experimental group were modeled with adjuvant arthritisand the normal group was given no treatment. After culture for 28 days, rats in the experimental group were sacrificed, and the third-generation FLSs were collected and randomly allocated into SOCS3 group, control group, and blank group. MTT assay was used for detecting cell viability, flow cytometry was used for analysis ofcell apoptosis, and enzyme-linked immunosorbent assay (ELISA) was used to determine levels of inflammatory factors (interleukin [IL]-2, interferon [IFN-γ] and tumor necrosis factor [TNF-α]). MTT assay showed that the optical density of the SOCS3 group was significantly higher than that of the control and blank groups. Flow cytometry showed that the apoptosis rate of FLSs in the SOCS3 group was significantly lower than that in the control and blank groups. The results of ELISA assay showed that the levels of IL-2, IFN-γ and TNF-α in the SOCS3 group were higher than those in the control and blank groups. Our study demonstrates that over-expression of SOCS3 promotes proliferation and inhibits apoptosis of FLSs in RA.
Collapse
Affiliation(s)
- Jia-Jing Zhu
- Department of Radiology, China-Japan Union Hospital of Jilin UniversityChangchun 130033, Jilin Province, P. R. China
| | - Zong-Qiang Wang
- Department of Medical, China-Japan Union Hospital of Jilin UniversityChangchun 130033, Jilin Province, P. R. China
| | - Li-Qi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin UniversityChangchun 130033, Jilin Province, P. R. China
| | - Gui-Feng Liu
- Department of Radiology, China-Japan Union Hospital of Jilin UniversityChangchun 130033, Jilin Province, P. R. China
| |
Collapse
|
41
|
Fan T, Zhang C, Zong M, Fan L. Hypoxia‑induced autophagy is inhibited by PADI4 knockdown, which promotes apoptosis of fibroblast‑like synoviocytes in rheumatoid arthritis. Mol Med Rep 2018; 17:5116-5124. [PMID: 29393388 PMCID: PMC5865976 DOI: 10.3892/mmr.2018.8501] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 09/12/2017] [Indexed: 12/15/2022] Open
Abstract
Impaired apoptosis of rheumatoid arthritis (RA)-fibroblast-like synoviocytes (FLS) is pivotal in the process of RA. Peptidyl arginine deiminase type IV (PADI4) is associated with autoantibody regulation via histone citrullination in RA. The present study aimed to investigate the role of PADI4 in the apoptosis of RA-FLS. FLS were isolated from patients with RA and a rat model. The effects of PADI4 on RA-FLS were investigated in vitro and in vivo. Hypoxia-induced autophagy was induced by 1% O2 and was detected by immunohistochemical and immunofluorescence analysis; in addition, apoptosis was detected by flow cytometry. RA-FLS obtained from RA rat model exhibited significant proliferation under severe hypoxia conditions. Hypoxia also significantly induced autophagy and elevated the expression of PADI4. Subsequently, short hairpin RNA-mediated PADI4 knockdown was demonstrated to significantly inhibit hypoxia-induced autophagy and promote apoptosis in RA-FLS. The results of these in vitro and in vivo studies suggested that PADI4 may be closely associated with hypoxia-induced autophagy, and the inhibition of hypoxia-induced autophagy by PADI4 knockdown may contribute to an increase in the apoptosis of RA-FLS.
Collapse
Affiliation(s)
- Tingting Fan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University Medical School, Shanghai 200120, P.R. China
| | - Changsong Zhang
- Clinical Oncology Laboratories, Changzhou Tumor Hospital, Medical College of Soochow University, Changzhou, Jiangsu 213032, P.R. China
| | - Ming Zong
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University Medical School, Shanghai 200120, P.R. China
| | - Lieying Fan
- Department of Clinical Laboratory, Shanghai East Hospital, Tongji University Medical School, Shanghai 200120, P.R. China
| |
Collapse
|
42
|
Ganesan R, Rasool M. Interleukin 17 regulates SHP-2 and IL-17RA/STAT-3 dependent Cyr61, IL-23 and GM-CSF expression and RANKL mediated osteoclastogenesis by fibroblast-like synoviocytes in rheumatoid arthritis. Mol Immunol 2017; 91:134-144. [PMID: 28898718 DOI: 10.1016/j.molimm.2017.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-17 predominately produced by the Th17 cells, plays a crucial role in the fibroblast-like synoviocytes (FLS) mediated disease process of rheumatoid arthritis (RA). IL-17 exerts its pathogenic effects in RA-FLS by IL-17/IL-17RA/STAT-3 signaling. Recent studies have shown that RA-FLS produces SHP-2, Cyr61, IL-23, GM-CSF and RANKL which results in worsening of the disease. However, whether IL-17/IL-17RA/STAT-3 signaling regulates SHP-2, Cyr61, IL-23, GM-CSF and RANKL expressions in RA-FLS remains unknown. In this study, IL-17 treatment dramatically induced the production of Cyr61, IL-23 and GM-CSF in FLS isolated from adjuvant induced arthritis (AA) rats. Conversely, IL-17 mediated production of Cyr61, IL-23 and GM-CSF was abrogated by knockdown of IL-17RA using a small interfering RNA or blockade of STAT-3 activation with S3I-201 in AA-FLS. Interestingly, IL-17 treatment noticeably increased the expression of IL-17RA and SHP-2 in AA-FLS. However, silencing of IL-17RA reversed the effect of IL-17 on the expression of IL-17RA and SHP-2 in AA-FLS. In addition, an increased number of TRAP-positive multinucleated cells were observed in a coculture system consisting of IL-17 treated AA-FLS and rat bone marrow derived monocytes/macrophages. Further, mechanistically we found that IL-17 upregulated RANKL expression in AA-FLS that was dependent on the IL-17/IL-17RA/STAT-3 signaling cascade. Knockdown of IL-17RA or inhibition of STAT-3 activation decreased the IL- 17 induced RANKL expression by AA-FLS and their osteoclastogenic potential. Taken together, our findings demonstrate that IL-17 regulates SHP-2 expression and IL-17RA/STAT-3 dependent production of Cyr61, IL-23, GM-CSF and RANKL in AA-FLS and may reveal a new insight into the pathogenesis of RA.
Collapse
Affiliation(s)
- Ramamoorthi Ganesan
- Immunopathology Lab, School of Bio Sciences and Technology, VIT University, Vellore 632 014, Tamilnadu, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Bio Sciences and Technology, VIT University, Vellore 632 014, Tamilnadu, India.
| |
Collapse
|
43
|
Jin J, Tao J, Gu X, Yu Z, Wang R, Zuo G, Li Q, Lv X, Miao D. P16 INK4a Deletion Ameliorated Renal Tubulointerstitial Injury in a Stress-induced Premature Senescence Model of Bmi-1 Deficiency. Sci Rep 2017; 7:7502. [PMID: 28790310 PMCID: PMC5548892 DOI: 10.1038/s41598-017-06868-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
To determine whether p16 INK4a deletion ameliorated renal tubulointerstitial injury by inhibiting a senescence-associated secretory phenotype (SASP) in Bmi-1-deficient (Bmi-1 -/-) mice, renal phenotypes were compared among 5-week-old Bmi-1 and p16 INK4a double-knockout, and Bmi-1 -/- and wild-type mice. Fifth-passage renal interstitial fibroblasts (RIFs) from the three groups were analyzed for senescence and proliferation. The effect of Bmi-1 deficiency on epithelial-to-mesenchymal transition (EMT) was examined in Bmi-1-knockdown human renal proximal tubular epithelial (HK2) cells, which were treated with concentrated conditioned medium (CM) from the fifth-passage renal interstitial fibroblasts (RIFs) of above three group mice or with exogenous TGF-β1. Our results demonstrated that p16 INK4a deletion largely rescued renal aging phenotypes caused by Bmi-1 deficiency, including impaired renal structure and function, decreased proliferation, increased apoptosis, senescence and SASP, DNA damage, NF-κB and TGF-β1/Smad signal activation, inflammatory cell infiltration, and tubulointerstitial fibrosis and tubular atrophy. P16 INK4a deletion also promoted proliferation, reduced senescence and SASP of RIFs and subsequently inhibited EMT of Bmi-1-knockdown HK2 cells. TGF-β1 further induced the EMT of Bmi-1-knockdown HK2 cells. Thus, p16 INK4a positive senescent cells would be a therapeutic target for preventing renal tubulointerstitial injury.
Collapse
MESH Headings
- Acute Kidney Injury/genetics
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/pathology
- Acute Kidney Injury/prevention & control
- Animals
- Cell Line, Transformed
- Cell Proliferation
- Cellular Senescence
- Coculture Techniques
- Culture Media, Conditioned/pharmacology
- Cyclin-Dependent Kinase Inhibitor p16/deficiency
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Gene Expression Regulation
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Mice
- Mice, Knockout
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/metabolism
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/prevention & control
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Polycomb Repressive Complex 1/deficiency
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Smad Proteins/genetics
- Smad Proteins/metabolism
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Jianliang Jin
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianguo Tao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Gu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhenzhen Yu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Rong Wang
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guoping Zuo
- Laboratory Centre for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, Jiangsu, 210029, China
| | - Xianhui Lv
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
44
|
Yin Y, Feng Y, Zhao H, Zhao Z, Yua H, Xu J, Che H. SIRT1 inhibits releases of HMGB1 and HSP70 from human umbilical vein endothelial cells caused by IL-6 and the serum from a preeclampsia patient and protects the cells from death. Biomed Pharmacother 2017; 88:449-458. [DOI: 10.1016/j.biopha.2017.01.087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/04/2017] [Accepted: 01/14/2017] [Indexed: 12/29/2022] Open
|
45
|
Mei ZG, Tan LJ, Wang JF, Li XL, Huang WF, Zhou HJ. Fermented Chinese formula Shuan-Tong-Ling attenuates ischemic stroke by inhibiting inflammation and apoptosis. Neural Regen Res 2017; 12:425-432. [PMID: 28469657 PMCID: PMC5399720 DOI: 10.4103/1673-5374.202946] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The fermented Chinese formula Shuan-Tong-Ling is composed of radix puerariae (Gegen), salvia miltiorrhiza (Danshen), radix curcuma (Jianghuang), hawthorn (Shanzha), salvia chinensis (Shijianchuan), sinapis alba (Baijiezi), astragalus (Huangqi), panax japonicas (Zhujieshen), atractylodes macrocephala koidz (Baizhu), radix paeoniae alba (Baishao), bupleurum (Chaihu), chrysanthemum (Juhua), rhizoma cyperi (Xiangfu) and gastrodin (Tianma), whose aqueous extract was fermented with lactobacillus, bacillus aceticus and saccharomycetes. Shuan-Tong-Ling is a formula used to treat brain diseases including ischemic stroke, migraine, and vascular dementia. Shuan-Tong-Ling attenuated H2O2-induced oxidative stress in rat microvascular endothelial cells. However, the potential mechanism involved in these effects is poorly understood. Rats were intragastrically treated with 5.7 or 17.2 mL/kg Shuan-Tong-Ling for 7 days before middle cerebral artery occlusion was induced. The results indicated Shuan-Tong-Ling had a cerebral protective effect by reducing infarct volume and increasing neurological scores. Shuan-Tong-Ling also decreased tumor necrosis factor-α and interleukin-1β levels in the hippocampus on the ischemic side. In addition, Shuan-Tong-Ling upregulated the expression of SIRT1 and Bcl-2 and downregulated the expression of acetylated-protein 53 and Bax. Injection of 5 mg/kg silent information regulator 1 (SIRT1) inhibitor EX527 into the subarachnoid space once every 2 days, four times, reversed the above changes. These results demonstrate that Shuan-Tong-Ling might benefit cerebral ischemia/reperfusion injury by reducing inflammation and apoptosis through activation of the SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Zhi-Gang Mei
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei Province, China.,Key Laboratory of Cardiovascular and Cerebrovascular Diseases Translational Medicine, China Three Gorges University, Yichang, Hubei Province, China
| | - Ling-Jing Tan
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei Province, China.,Key Laboratory of Cardiovascular and Cerebrovascular Diseases Translational Medicine, China Three Gorges University, Yichang, Hubei Province, China
| | - Jin-Feng Wang
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei Province, China
| | - Xiao-Li Li
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei Province, China
| | - Wei-Feng Huang
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei Province, China
| | - Hua-Jun Zhou
- Institute of Neurology, the First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei Province, China
| |
Collapse
|