101
|
Remodeling articular immune homeostasis with an efferocytosis-informed nanoimitator mitigates rheumatoid arthritis in mice. Nat Commun 2023; 14:817. [PMID: 36781864 PMCID: PMC9925448 DOI: 10.1038/s41467-023-36468-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/02/2023] [Indexed: 02/15/2023] Open
Abstract
Massive intra-articular infiltration of proinflammatory macrophages is a prominent feature of rheumatoid arthritis (RA) lesions, which are thought to underlie articular immune dysfunction, severe synovitis and ultimately joint erosion. Here we report an efferocytosis-informed nanoimitator (EINI) for in situ targeted reprogramming of synovial inflammatory macrophages (SIMs) that thwarts their autoimmune attack and reestablishes articular immune homeostasis, which mitigates RA. The EINI consists of a drug-based core with an oxidative stress-responsive phosphatidylserine (PtdSer) corona and a shell composed of a P-selectin-blocking motif, low molecular weight heparin (LMWH). When systemically administered, the LMWH on the EINI first binds to P-selectin overexpressed on the endothelium in subsynovial capillaries, which functions as an antagonist, disrupting neutrophil synovial trafficking. Due to the strong dysregulation of the synovial microvasculature, the EINI is subsequently enriched in the joint synovium where the shell is disassembled upon the reactive oxygen species stimulation, and PtdSer corona is then exposed. In an efferocytosis-like manner, the PtdSer-coroneted core is in turn phagocytosed by SIMs, which synergistically terminate SIM-initiated pathological cascades and serially reestablish intra-articular immune homeostasis, conferring a chondroprotective effect. These findings demonstrate that SIMs can be precisely remodeled via the efferocytosis-mimetic strategy, which holds potential for RA treatment.
Collapse
|
102
|
Jing W, Liu C, Su C, Liu L, Chen P, Li X, Zhang X, Yuan B, Wang H, Du X. Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Front Immunol 2023; 14:1107670. [PMID: 36845127 PMCID: PMC9948260 DOI: 10.3389/fimmu.2023.1107670] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, pannus formation, and bone and cartilage damage. It has a high disability rate. The hypoxic microenvironment of RA joints can cause reactive oxygen species (ROS) accumulation and mitochondrial damage, which not only affect the metabolic processes of immune cells and pathological changes in fibroblastic synovial cells but also upregulate the expression of several inflammatory pathways, ultimately promoting inflammation. Additionally, ROS and mitochondrial damage are involved in angiogenesis and bone destruction, thereby accelerating RA progression. In this review, we highlighted the effects of ROS accumulation and mitochondrial damage on inflammatory response, angiogenesis, bone and cartilage damage in RA. Additionally, we summarized therapies that target ROS or mitochondria to relieve RA symptoms and discuss the gaps in research and existing controversies, hoping to provide new ideas for research in this area and insights for targeted drug development in RA.
Collapse
Affiliation(s)
- Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chenghong Su
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangjun Li
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
103
|
Stimulus-responsive and dual-target DNA nanodrugs for rheumatoid arthritis treatment. Int J Pharm 2023; 632:122543. [PMID: 36572263 DOI: 10.1016/j.ijpharm.2022.122543] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Tumor necrosis factor receptor-1 (TNFR1) and DEK are closely associated with the development of rheumatoid arthritis (RA). Taking advantage of the high adenosine triphosphate (ATP) in RA microenvironment and the interactions of DNA aptamers with their targets, an ATP-responsive DNA nanodrug was constructed that simultaneously targets TNFR1 and DEK for RA therapy. To this end, DEK target aptamer DTA and TNFR1 target aptamer Apt1-67 were equipped with sticky ends to hybridize with ATP aptamer (AptATP) and fabricated DNA nanodrug DAT. Our results showed that DAT was successfully prepared with good stability. In the presence of ATP, DAT was disassembled, resulting in the release of DTA and Apt1-67. In vitro studies demonstrated that DAT was superior to the non-responsive DNA nanodrug TD-3A3T in terms of anti-inflammation activity and ATP was inevitable to maximize the anti-inflammation ability of DAT. The superior efficacy of DAT is attributed to the more potent inhibition of caspase-3 and NETs formation. In vivo results further confirmed the anti-RA efficacy of DAT, whereas the administration routes (intravenous injection and transdermal administration via microneedles) did not cause significant differences. Overall, the present study supplies an intelligent strategy for RA therapy and explores a promising administration route for future clinical medication of RA patients.
Collapse
|
104
|
Zhang RD, Chen C, Wang P, Fang Y, Jiang LQ, Fang X, Zhao Y, Ni J, Wang DG, Pan HF. Air pollution exposure and auto-inflammatory and autoimmune diseases of the musculoskeletal system: a review of epidemiologic and mechanistic evidence. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023:10.1007/s10653-023-01495-x. [PMID: 36735155 DOI: 10.1007/s10653-023-01495-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Auto-inflammatory and autoimmune diseases of the musculoskeletal system can be perceived as a spectrum of rheumatic diseases, with the joints and connective tissues are eroded severely that progressively develop chronic inflammation and lesion. A wide range of risk factors represented by genetic and environmental factors have been uncovered by population-based surveys and experimental studies. Lately, the exposure to air pollution has been found to be potentially involved in the mechanisms of occurrence or development of such diseases, principally manifest in oxidative stress, local and systemic inflammation, and epigenetic modifications, as well as the mitochondrial dysfunction, which has been reported to participate in the intermediate links. The lungs might serve as a starting area of air pollutants, which would cause oxidative stress-induced bronchial-associated lymphoid tissue (iBALT) to further to influence T, B cells, and the secretion of pro-inflammatory cytokines. The binding of aromatic hydrocarbon receptor (AhR) to the corresponding contaminant ligands tends to regulate the reaction of Th17 and Tregs. Furthermore, air pollution components might spur on immune and inflammatory responses by damaging mitochondria that could interact with and exacerbate oxidative stress and pro-inflammatory cytokines. In this review, we focused on the association between air pollution and typical auto-inflammatory and autoimmune diseases of the musculoskeletal system, mainly including osteoarthritis (OA), rheumatoid arthritis (RA), spondyloarthritis (SpA) and juvenile idiopathic arthritis (JIA), and aim to collate the mechanisms involved and the potential channels. A complete summary and in-depth understanding of the autoimmune and inflammatory effects of air pollution exposure should hopefully contribute new perspectives on how to formulate better public health policies to alleviate the adverse health effects of air pollutants.
Collapse
Affiliation(s)
- Ruo-Di Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Cong Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Peng Wang
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Yang Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Xi Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Yan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - De-Guang Wang
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
- Department of Nephrology, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China.
- Institute of Kidney Disease, Inflammation and Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
105
|
Barker BE, Hanlon MM, Marzaioli V, Smith CM, Cunningham CC, Fletcher JM, Veale DJ, Fearon U, Canavan M. The mammalian target of rapamycin contributes to synovial fibroblast pathogenicity in rheumatoid arthritis. Front Med (Lausanne) 2023; 10:1029021. [PMID: 36817783 PMCID: PMC9936094 DOI: 10.3389/fmed.2023.1029021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Objectives The mammalian target of Rapamycin (mTOR) is a metabolic master regulator of both innate and adaptive immunity; however, its exact role in stromal cell biology is unknown. In this study we explored the role of the mTOR pathway on Rheumatoid Arthritis synovial fibroblast (RASF) metabolism and activation and determined if crosstalk with the Hippo-YAP pathway mediates their effects. Methods Primary RA synovial fibroblasts (RASF) were cultured with TNFα alone or in combination with the mTOR inhibitor Rapamycin or YAP inhibitor Verteporfin. Chemokine production, matrix metalloproteinase (MMP) production, and adhesion marker expression were quantified by real-time PCR, ELISA, and/or Flow Cytometry. Invasion assays were performed using Transwell invasion chambers, while wound repair assays were used to assess RASF migration. Cellular bioenergetics was assessed using the Seahorse XFe96 Analyzer. Key metabolic genes (GLUT-1, HK2, G6PD) were measured using real-time PCR. Reanalysis of RNA-Seq analysis was performed on RA (n = 151) and healthy control (HC) (n = 28) synovial tissue biopsies to detect differential gene and pathway expression. The expression of YAP was measured by Western Blot. Results Transcriptomic analysis of healthy donor and RA synovial tissue revealed dysregulated expression of several key components of the mTOR pathway in RA. Moreover, the expression of phospho-ribosomal protein S6 (pS6), the major downstream target of mTOR is specifically increased in RA synovial fibroblasts compared to healthy tissue. In the presence of TNFα, RASF display heightened phosphorylation of S6 and are responsive to mTOR inhibition via Rapamycin. Rapamycin effectively alters RASF cellular bioenergetics by inhibiting glycolysis and the expression of rate limiting glycolytic enzymes. Furthermore, we demonstrate a key role for mTOR signaling in uniquely mediating RASF migratory and invasive mechanisms, which are significantly abrogated in the presence of Rapamycin. Finally, we report a significant upregulation in several genes involved in the Hippo-YAP pathway in RA synovial tissue, which are predicted to converge with the mTOR pathway. We demonstrate crosstalk between the mTOR and YAP pathways in mediating RASF invasive mechanism whereby Rapamycin significantly abrogates YAP expression and YAP inhibition significantly inhibits RASF invasiveness. Conclusion mTOR drives pathogenic mechanisms in RASF an effect which is in part mediated via crosstalk with the Hippo-YAP pathway.
Collapse
Affiliation(s)
- Brianne E. Barker
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,Translational Immunopathology, School of Biochemistry & Immunology and School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Megan M. Hanlon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Viviana Marzaioli
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Conor M. Smith
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Clare C. Cunningham
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Douglas J. Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Mary Canavan
- Translational Immunopathology, School of Biochemistry & Immunology and School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,*Correspondence: Mary Canavan,
| |
Collapse
|
106
|
Dai Z, Xia C, Zhao T, Wang H, Tian H, Xu O, Zhu X, Zhang J, Chen P. Platelet-derived extracellular vesicles ameliorate intervertebral disc degeneration by alleviating mitochondrial dysfunction. Mater Today Bio 2023; 18:100512. [PMID: 36536658 PMCID: PMC9758573 DOI: 10.1016/j.mtbio.2022.100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction causes the production of reactive oxygen species (ROS) and oxidative damage, and oxidative stress and inflammation are considered key factors causing intervertebral disc degeneration (IVDD). Thus, restoring the mitochondrial dysfunction is an attractive strategy for treating IVDD. Platelet-derived extracellular vesicles (PEVs) are nanoparticles that target inflammation. Moreover, the vesicles produced by platelets (PLTs) have considerable anti-inflammatory effects. We investigate the use of PEVs as a therapeutic strategy for IVDD in this study. We extract PEVs and evaluate their properties; test their effects on H2O2-induced oxidative damage of nucleus pulposus (NP) cells; verify the role of PEVs in repairing H2O2-induced cellular mitochondrial dysfunction; and demonstrate the therapeutic effects of PEVs in a rat IVDD model. The results confirm that PEVs can restore impaired mitochondrial function, reduce oxidative stress, and restore cell metabolism by regulating the sirtuin 1 (SIRT1)-peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α)-mitochondrial transcription factor A (TFAM) pathway; in rat models, PEVs retard the progression of IVDD. Our results demonstrate that the injection of PEVs can be a promising strategy for treating patients with IVDD.
Collapse
Affiliation(s)
- Zhanqiu Dai
- Department of Orthopaedics, The Second Affiliated Hospital of Bengbu Medical College, Anhui, China
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical College People's Hospital, Hangzhou, Zhejiang, China
| | - Chen Xia
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical College People's Hospital, Hangzhou, Zhejiang, China
| | - Tingxiao Zhao
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical College People's Hospital, Hangzhou, Zhejiang, China
| | - Haoli Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Hongsen Tian
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Ouyuan Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xunbin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Bengbu Medical College, Anhui, China
| | - Jun Zhang
- Department of Spine Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical College People's Hospital, Hangzhou, Zhejiang, China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, China
| |
Collapse
|
107
|
Han J, Wang J, Wang Y, Zhu Z, Zhang S, Wu B, Meng M, Zhao J, Wang D. Sesquiterpene lactones-enriched fractions from Xanthium mongolicum Kitag alleviate RA by regulating M1 macrophage polarization via NF-κB and MAPK signaling pathway. Front Pharmacol 2023; 14:1104153. [PMID: 36778009 PMCID: PMC9909009 DOI: 10.3389/fphar.2023.1104153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction: Rheumatoid arthritis (RA) is a chronic autoimmune disease, characterized by activated M1-like macrophage in the joint. Xanthium mongolicum Kitag (X. mongolicum) is a traditional medicinal plant that has long been used to treat RA and other immune diseases in China. Methods: Fractions of X. mongolicum were separated based on polarity. Anti-RA activity of the fractions were screened by LPS-stimulated RAW264.7 macrophage in vitro. The major active compounds were identified by UPLC-MS and quantified by HPLC. The anti-RA effects of the active fraction was evaluated in complete freund's adjuvant (CFA)-induced arthritis and collagen-induced arthritis (CIA) mouse models in vivo and LPS-stimulated macrophage in vitro. Results: Sesquiterpene lactones-enriched fraction from X. mongolicum (SL-XM) exhibited the strongest anti-RA activity among all components in vitro. Five major constituents i.e., Xanthinosin (1), Xanthatin (2), Mogolide D (3), Mogolide E (4), and Mogolide A (5) were identified as major compounds of SL-XM. SL-XM ameliorated symptoms of CFA and CIA induced arthritis mice model. Furthermore, SL-XM treatment inhibited LPS-induced M1 macrophages polarization. In addition, SL-XM inhibited the phosphorylation of NF-κB and MAPK signaling pathways in LPS-induced macrophage and CIA-challenged mice. Discussion: The main anti-RA active fraction of X. mongolicum may be the Sesquiterpene lactones, which includes five key compounds. SL-XM may exert its anti-RA effect by suppressing M1 macrophage polarization via the NF-κB and MAPK signaling pathway.
Collapse
Affiliation(s)
- Jing Han
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China,*Correspondence: Jing Han, ; Jianning Zhao, ; Dongsheng Wang,
| | - Jingwen Wang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yicun Wang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Zhiqi Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siwang Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bingrong Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingsong Meng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China,*Correspondence: Jing Han, ; Jianning Zhao, ; Dongsheng Wang,
| | - Dongsheng Wang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China,*Correspondence: Jing Han, ; Jianning Zhao, ; Dongsheng Wang,
| |
Collapse
|
108
|
Greeck VB, Williams SK, Haas J, Wildemann B, Fairless R. Alterations in Lymphocytic Metabolism-An Emerging Hallmark of MS Pathophysiology? Int J Mol Sci 2023; 24:ijms24032094. [PMID: 36768415 PMCID: PMC9917089 DOI: 10.3390/ijms24032094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterised by acute inflammation and subsequent neuro-axonal degeneration resulting in progressive neurological impairment. Aberrant immune system activation in the periphery and subsequent lymphocyte migration to the CNS contribute to the pathophysiology. Recent research has identified metabolic dysfunction as an additional feature of MS. It is already well known that energy deficiency in neurons caused by impaired mitochondrial oxidative phosphorylation results in ionic imbalances that trigger degenerative pathways contributing to white and grey matter atrophy. However, metabolic dysfunction in MS appears to be more widespread than the CNS. This review focuses on recent research assessing the metabolism and mitochondrial function in peripheral immune cells of MS patients and lymphocytes isolated from murine models of MS. Emerging evidence suggests that pharmacological modulation of lymphocytic metabolism may regulate their subtype differentiation and rebalance pro- and anti-inflammatory functions. As such, further understanding of MS immunometabolism may aid the identification of novel treatments to specifically target proinflammatory immune responses.
Collapse
Affiliation(s)
- Viktoria B. Greeck
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sarah K. Williams
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jürgen Haas
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Brigitte Wildemann
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
109
|
Zhang HJ, Li JY, Wang C, Zhong GQ. Microvesicles with mitochondrial content are increased in patients with sepsis and associated with inflammatory responses. World J Clin Cases 2023; 11:342-356. [PMID: 36686348 PMCID: PMC9850980 DOI: 10.12998/wjcc.v11.i2.342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/03/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Endothelial activation plays an important role in sepsis-mediated inflammation, but the triggering factors have not been fully elucidated. Microvesicles carrying mitochondrial content (mitoMVs) have been implicated in several diseases and shown to induce endothelial activation.
AIM To explore whether mitoMVs constitute a subset of MVs isolated from plasma of patients with sepsis and contribute to endothelial activation.
METHODS MVs were isolated from human plasma and characterized by confocal microscopy and flow cytometry. Proinflammatory cytokines, including interleukin (IL)-6, IL-8 and tumour necrosis factor (TNF)-α, and soluble vascular cell adhesion molecule (sVCAM)-1 were detected by ELISA. Human umbilical vein endothelial cells (HUVECs) were stimulated with the circulating MVs to evaluate their effect on endothelial activation.
RESULTS MitoMVs were observed in plasma from patients with sepsis. Compared with those in healthy controls, expression of MVs, mitoMVs, proinflammatory cytokines and sVCAM-1 was increased. The number of mitoMVs was positively associated with TNF-α and sVCAM-1. In vitro, compared with MVs isolated from the plasma of healthy controls, MVs isolated from the plasma of patients with sepsis induced expression of OAS2, RSAD2, and CXCL10 in HUVECs. MitoMVs were taken up by HUVECs, and sonication of MVs significantly reduced the uptake of mitoMVs by HUVECs and expression of the above three type I IFN-dependent genes.
CONCLUSION MitoMVs are increased in the plasma of patients with sepsis, which induces elevated expression of type I IFN-dependent genes. This suggests that circulating mitoMVs activate the type I IFN signalling pathway in endothelial cells and lead to endothelial activation.
Collapse
Affiliation(s)
- Hai-Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, Hunan Province, China
| | - Jin-Yi Li
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541000, Guangxi Zhuang Autonomous Region, China
| | - Chao Wang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Guo-Qiang Zhong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
110
|
Hypoxia and TNF-α Synergistically Induce Expression of IL-6 and IL-8 in Human Fibroblast-like Synoviocytes via Enhancing TAK1/NF-κB/HIF-1α Signaling. Inflammation 2023; 46:912-924. [PMID: 36607540 DOI: 10.1007/s10753-022-01779-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023]
Abstract
Hypoxia and increased levels of inflammatory cytokines in the joints are characteristics of rheumatoid arthritis (RA). However, the effects of hypoxia and tumor necrosis factor-α (TNF-α) on interleukin (IL)-6 and IL-8 production on fibroblast-like synoviocytes (FLSs) remain to be clarified. This study aimed to explore how hypoxia and TNF-α affect the expression of IL-6 and IL-8 in human FLSs isolated from RA patients. Hypoxia or TNF-α treatment alone significantly increased the expression and promoter activity of IL-6, IL-8, and hypoxia-inducible factor-1α (HIF-1α). Treatment of hypoxic FLSs with TNF-α further significantly elevated the expression of these cytokines and enhanced promoter activity of HIF-1α, which was abrogated by treatment with the HIF-1α inhibitor YC-1. Similarly, TNF-α alone elevated the phosphorylation and promoter activity of nuclear factor-κBp65 (NF-κBp65) in the FLSs. These effects were further enhanced by the combined treatment of hypoxia and TNFα but were attenuated by the NF-κB inhibitor BAY11-7082. NF-κB-p65 inhibition decreased the effect of TNF-α on HIF-1α upregulation in the FLSs in response to hypoxia. The combination of hypoxia and TNF-α also significantly upregulated transforming growth factor-β-activated kinase 1 (TAK1) expression, and silencing TAK1 dramatically decreased NF-κB-p65, HIF-1α, IL-6, and IL-8 expression under the same conditions. Our results indicate that hypoxia and TNF-α synergistically increase IL-6 and IL-8 expression in human FLSs via enhancing TAK1/NF-κB/HIF-1α signaling.
Collapse
|
111
|
Ding C, Ni L, Liu Q, Zhou C, Wang G, Chu PK, Wu Z. Cold air plasma improving rheumatoid arthritis via mitochondrial apoptosis pathway. Bioeng Transl Med 2023; 8:e10366. [PMID: 36684093 PMCID: PMC9842019 DOI: 10.1002/btm2.10366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/03/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023] Open
Abstract
Rheumatoid arthritis (RA) has plagued physicians and patients for years due to the lack of targeted treatment. In this study, inspired by the commonality between rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) and cancer cells, the therapeutic effects of cold air plasma (CAP) on RA are studied systematically and thoroughly. In/ex vivo results show that CAP with the proper dosage significantly relieves symptoms including synovial hyperplasia, inflammatory infiltration, and angiogenesis and eliminates the root cause by triggering the self-antioxidant capability of the surrounding tissue. The mechanism on the molecular and cellular level is also revealed that the spontaneous reactive oxygen species (ROS) cascade induces the mitochondrial apoptosis pathway on RA-FLS. This study reveals a new strategy for targeted treatment of RA and the mechanistic study provides the theoretical foundation for future development of plasma medicine.
Collapse
Affiliation(s)
- Chengbiao Ding
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaHefeiChina
- Department of Rehabilitation MedicineThe Second Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Leying Ni
- Department of Rehabilitation MedicineThe Second Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Qi Liu
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaHefeiChina
| | - Chenxu Zhou
- Department of Rehabilitation MedicineThe Second Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Guomin Wang
- Department of PhysicsCity University of Hong KongKowloon, Hong KongChina
- Department of Biomedical EngineeringCity University of Hong KongKowloon, Hong KongChina
- Department of Materials Science and EngineeringCity University of Hong KongKowloon, Hong KongChina
| | - Paul K. Chu
- Department of PhysicsCity University of Hong KongKowloon, Hong KongChina
- Department of Biomedical EngineeringCity University of Hong KongKowloon, Hong KongChina
- Department of Materials Science and EngineeringCity University of Hong KongKowloon, Hong KongChina
| | - Zhengwei Wu
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaHefeiChina
- CAS Key Laboratory of Geospace EnvironmentUniversity of Science and Technology of ChinaHefeiChina
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
112
|
Yu B, Chen Y, Chen E, Zuo F, Yuan Y, Zhao X, Xiao C. LncRNA RNA XIST binding to GATA1 contributes to rheumatoid arthritis through its effects on proliferation of synovial fibroblasts and angiogenesis via regulation of CCN6. Mol Immunol 2023; 153:200-211. [PMID: 36542956 DOI: 10.1016/j.molimm.2022.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
This study explored the role of the long non-coding RNA (lncRNA) XIST (X-inactive specific transcript) as a driver of RA pathogenesis, with a particular focus on the ability of this lncRNA to interact with GATA1 and CCN6. The GSE83147and GSE181614 datasets were downloaded for analysis. XIST and CCN6 expression were assessed in synovial fibroblasts (SFs) and in both normal cartilage samples and those from RA patients, with the relationship between XIST and CCN6 additionally being examined. XIST and CCN6 were respectively knocked down or overexpressed in SFs to establish their regulatory roles in these cells in the context of RA. Further studies of the regulatory interplay between XIST, GATA1, and CCN6 were then performed through RNA immunoprecipitation, RNA pull-down, gain-of-function, loss-of-function, and luciferase reporter assays. In addition, RA model rats were established and used to measure the production of TNF-α, IL-6, and IL-8 and to subject tissues from these animals to histopathological examination. RA patient synovial tissues and SFs exhibited XIST and CCN6 upregulation. The knockdown of XIST suppressed SF migratory, proliferative, invasive, and angiogenic activity, while CCN6 knockdown partially reversed the ability of XIST to influence these phenotypic outcomes in vitro and in vivo. XIST bound to GATA1 within SFs, thus promoting enhanced CCN6 transcription. Knocking down XIST alleviated RA-related pathological damage, synovial injury, and inflammatory response induction in rats. The binding of XIST to GATA1 leads to CCN6 upregulation, driving RA pathogenesis by altering SF proliferation and angiogenic activity, suggesting that this pathway may represent a viable target for therapeutic intervention.
Collapse
Affiliation(s)
- Beijia Yu
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Yong Chen
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Ensheng Chen
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Fangfang Zuo
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Yi Yuan
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Xiaofeng Zhao
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| | - Changhong Xiao
- the Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, China.
| |
Collapse
|
113
|
HIF-1α promotes paraquat induced acute lung injury and implicates a role NF-κB and Rac2 activity. Toxicology 2023; 483:153388. [PMID: 36462643 DOI: 10.1016/j.tox.2022.153388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/04/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
Paraquat (PQ) is a bipyridine herbicide and oral exposure is the main way of PQ exposure with a very high mortality. At present, it is believed that large number of oxygen free radicals are generated and cause lipid peroxidation of tissue and organ cell membranes after PQ is absorbed. PQ exposure could cause multiple organ dysfunction, among which acute lung injury is the most common and most serious. However, its specific mechanism is still unclear. In this study, the C57BL/6J mouse (alveolar epithelial cell-specific knockout HIF-1α) model of acute lung injury (40 mg/kg PQ) at several time pointes and a model of acute type II alveolar epithelial cell (A549, 800 μM PQ) injury constructed. The oxidative stress (ROS, MDA) and inflammatory response (IL-1β, IL-6, TNF-α) were significantly inhibited in the alveolar epithelial cell-specific knockout of HIF-1α mice and siRNA technology to inhibit HIF-1α in alveolar epithelial cells. Further proteomic analysis showed that the expression of Rac2 protein, which is closely related to oxidative stress, was significantly increased after PQ exposure. And the inhibition of Rac2 expression in vitro significantly alleviated PQ-induced oxidative stress and inflammatory response. The expression of Rac2 protein was regulated by HIF-1α. The above suggests that HIF-1α may promote oxidative stress and inflammatory response in alveolar epithelial cells by regulating the expression of Rac2, and then participate in the promotion of PQ exposure-induced acute lung injury.
Collapse
|
114
|
Wang J, Chen Q, Sheng R, Li P, Liu P, Luo J, Zhong Z, Xu S. Integration of transdermal chemistry and network pharmacology to decipher the mechanism of ShexiangZhuifeng analgesic plaster to treat rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154507. [PMID: 36334391 DOI: 10.1016/j.phymed.2022.154507] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/04/2022] [Accepted: 10/16/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Transdermal patches are an effective form of treatment for rheumatoid arthritis (RA), and they have a number of benefits, including patient compliance, accessibility, and low systemic toxicity. ShexiangZhuifeng Analgesic Plaster (SZAP), a patch made up of many traditional medicines, has been successfully utilized in numerous clinical trials to treat RA. However, information about anti-RA processes and transdermal active components is still emerging. PURPOSE Our objectives were to identify the transdermal active components of SZAP and investigate its anti-RA mechanisms, primarily focused on joint inflammation. METHODS The collagen-induced arthritis (CIA) rats were created first, and then the arthritis score, Paw thickness, and morphology feature of joint synovial were assessed after 7 days of therapy with SZAP. Moreover, the Franz diffusion cell and UPLC-MS technologies were combined to identify and measure the transdermal active ingredients of SZAP. Furthermore, network pharmacology was utilized to anticipate the putative the mechanism of SZAP for treating RA. Finally, the results of network pharmacology were validated using LPS-induced RAW 264.7 cells and CIA rats. RESULTS SZAP significantly reduced paw thickness, arthritic score and pathological characteristics of joint synovitis in (CIA) rats. Additionally, 12 transdermal active components of SZAP were identified, and network pharmacology prediction results suggested that SZAP may alleviate joint synovial inflammation by blocking the Akt/mTOR/HIF-1 pathway. Our investigations' findings demonstrated that SZAP dramatically reduced the concentrations of excess cytokines (IL6, VEGF, and TNF-α), as well as the protein overexpression of the AKT/mTOR/HIF- pathway (HIF-1, p-AKT, and p-mTOR), whereas its anti-inflammation effect was reversed once AKT or mTOR was activated. CONCLUSION By blocking the AKT/mTOR/HIF-1 pathway, SZAP can lessen the release of inflammatory mediators, which reduces joint synovial inflammation associated with RA. The pharmacological evaluation of TCM transdermal drug delivery formulations like SZAP may be amenable to the integration of transdermal chemistry and network pharmacology approaches.
Collapse
Affiliation(s)
- Jie Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Qi Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Ruilin Sheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Ping Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Panwang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Jie Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Zhanqiong Zhong
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Shijun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan, 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| |
Collapse
|
115
|
Ospelt C. Site of invasion revisited: epigenetic drivers of joint destruction in RA. Ann Rheum Dis 2022; 82:734-739. [PMID: 36585124 DOI: 10.1136/ard-2022-222554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022]
Abstract
New analytical methods and the increasing availability of synovial biopsies have recently provided unprecedented insights into synovial activation in general and synovial fibroblast (SF) biology in particular. In the course of this development, SFs have become one of the most rapidly evolving and exciting fields of rheumatoid arthritis (RA) research. While their active role in the invasion of RA synovium into cartilage has long been studied, recent studies have brought new aspects of their heterogeneity and propagation in RA. This review integrates old and new evidence to give an overview picture of the processes active at the sites of invasive synovial tissue growth in RA.
Collapse
Affiliation(s)
- Caroline Ospelt
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
116
|
Advances in Human Mitochondria-Based Therapies. Int J Mol Sci 2022; 24:ijms24010608. [PMID: 36614050 PMCID: PMC9820658 DOI: 10.3390/ijms24010608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Mitochondria are the key biological generators of eukaryotic cells, controlling the energy supply while providing many important biosynthetic intermediates. Mitochondria act as a dynamic, functionally and structurally interconnected network hub closely integrated with other cellular compartments via biomembrane systems, transmitting biological information by shuttling between cells and tissues. Defects and dysregulation of mitochondrial functions are critically involved in pathological mechanisms contributing to aging, cancer, inflammation, neurodegenerative diseases, and other severe human diseases. Mediating and rejuvenating the mitochondria may therefore be of significant benefit to prevent, reverse, and even treat such pathological conditions in patients. The goal of this review is to present the most advanced strategies using mitochondria to manage such disorders and to further explore innovative approaches in the field of human mitochondria-based therapies.
Collapse
|
117
|
Rashid MM, Corbin BA, Jella P, Ortiz CJ, Samee MAH, Pautler RG, Allen MJ. Systemic Delivery of Divalent Europium from Ligand Screening with Implications to Direct Imaging of Hypoxia. J Am Chem Soc 2022; 144:23053-23060. [PMID: 36475663 PMCID: PMC9782726 DOI: 10.1021/jacs.2c10373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia is a hallmark of many diseases, including cancer, arthritis, heart and kidney diseases, and diabetes, and it is often associated with disease aggressiveness and poor prognosis. Consequently, there is a critical need for imaging hypoxia in a noninvasive and direct way to diagnose, stage, and monitor the treatment and development of new therapies for these diseases. Eu-containing contrast agents for magnetic resonance imaging have demonstrated potential for in vivo imaging of hypoxia via changes in metal oxidation state from +2 to +3, but rapid oxidation in blood limits EuII-containing complexes to studies compatible with direct injection to sites. Here, we report a new EuII-containing complex that persists in oxygenated environments and is capable of persisting in blood long enough for imaging by magnetic resonance imaging. We describe the screening of a library of ligands that led to the discovery of the complex as well as a pH-dependent mechanism that hinders oxidation to enable usefulness in vivo. These studies of the first divalent lanthanide complex that persists in oxygenated solutions open the door to the use of EuII-based contrast agents for imaging hypoxia in a wide range of diseases.
Collapse
Affiliation(s)
- Md Mamunur Rashid
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, USA
| | - Brooke A. Corbin
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, USA
| | - Pavan Jella
- Department of Radiology, Wayne State University, Detroit, Michigan 48201, USA
| | - Caitlyn J. Ortiz
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Md. Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Robia G. Pautler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Matthew J. Allen
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, USA
| |
Collapse
|
118
|
Wan L, Liu J, Huang C, Zhu Z, Li F, Sun G, Wang K, Li S, Ma X, Chen X, Yuan W. Role of m6A modification and novel circ_0066715/ miR-486-5p/ ETS1 axis in rheumatoid arthritis macrophage polarization progression. Aging (Albany NY) 2022; 14:10009-10026. [PMID: 36541909 PMCID: PMC9831719 DOI: 10.18632/aging.204439] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic disease dominated by inflammatory synovitis. RA synovial macrophages tend undergo M1-type macrophage polarization. Then, polarized M1-type macrophages secrete abundant pro-inflammatory cytokines, causing joint and cartilage destruction. N6-methyladenosine (m6A) methylation modification, circular RNA (circRNA), microRNA (miRNA), messenger RNA (mRNA), etc. are involved in the inflammatory response of RA. We found that there is an imbalance of inflammatory polarization in RA, which is manifested by a sharp increase in inflammatory markers and a high inflammatory response. Here, we show that RA was closely associated with low expression of circ_0066715. The overexpression of circ_0066715 significantly increased the ETS1 levels in RA-FLS cells, decreased cytokine secretion by M1-type macrophages, elevated M2-type cytokines, and inhibited FLS proliferation. Interestingly, the overexpression of miR-486-5p significantly suppressed the attenuation of the cell function and the effect on M1 macrophage polarization caused by circ_0066715 positive expression. WTAP may be involved in the methylation process of ETS1 in RA. ETS1 m6A methylation levels were altered upon WTAP intervention. The overexpression or interference of circ_0066715 decreased or increased WTAP expression. Our findings provide a novel circRNA/miRNA/mRNA regulatory axis and m6A regulatory mechanism involved in the process of RA macrophage polarization, thereby providing a powerful diagnostic and therapeutic strategy for RA treatment.
Collapse
Affiliation(s)
- Lei Wan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China,Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China
| | - Jian Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China,Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China
| | - Chuanbing Huang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ziheng Zhu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Fangze Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Guanghan Sun
- College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Kun Wang
- Key Laboratory of Xin’an Medical Education Ministry, Hefei 230038, China,College of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Shu Li
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ximeng Ma
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xi Chen
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| | - Wang Yuan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230038, China
| |
Collapse
|
119
|
Reoxygenation Modulates the Adverse Effects of Hypoxia on Wound Repair. Int J Mol Sci 2022; 23:ijms232415832. [PMID: 36555485 PMCID: PMC9781139 DOI: 10.3390/ijms232415832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022] Open
Abstract
Hypoxia is a major stressor and a prominent feature of pathological conditions, such as bacterial infections, inflammation, wounds, and cardiovascular defects. In this study, we investigated whether reoxygenation has a protective effect against hypoxia-induced acute injury and burn using the C57BL/6 mouse model. C57BL/6 mice were exposed to hypoxia and treated with both acute and burn injuries and were in hypoxia until wound healing. Next, C57BL/6 mice were exposed to hypoxia for three days and then transferred to normoxic conditions for reoxygenation until wound healing. Finally, skin wound tissue was collected to analyze healing-related markers, such as inflammation, vascularization, and collagen. Hypoxia significantly increased inflammatory cell infiltration and decreased vascular and collagen production, and reoxygenation notably attenuated hypoxia-induced infiltration of inflammatory cells, upregulation of pro-inflammatory cytokine levels (IL-6 and TNF-α) in the wound, and remission of inflammation in the wound. Immunofluorescence analysis showed that reoxygenation increased the expression of the angiogenic factor α-SMA and decreased ROS expression in burn tissues compared to hypoxia-treated animals. Moreover, further analysis by qPCR showed that reoxygenation could alleviate the expression of hypoxic-induced inflammatory markers (IL-6 and TNF), increase angiogenesis (SMA) and collagen synthesis (Col I), and thus promote wound healing. It is suggested that oxygen can be further evaluated in combination with oxygen-releasing materials as a supplementary therapy for patients with chronic hypoxic wounds.
Collapse
|
120
|
Yin Y, Xie Y, Ge W, Li Y. Creeping fat formation and interaction with intestinal disease in Crohn's disease. United European Gastroenterol J 2022; 10:1077-1084. [PMID: 36507842 PMCID: PMC9752293 DOI: 10.1002/ueg2.12349] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Creeping fat (CrF), also known as fat wrapping, is a significant disease characteristic of Crohn's disease (CD). The transmural inflammation impairs intestinal integrity and facilitates bacteria translocation, aggravating immune response. CrF is a rich source of pro-inflammatory and pro-fibrotic cytokines with complex immune microenvironment. The inflamed and stricturing intestine is often wrapped by CrF, and CrF is associated with greater severity of CD. The large amount of innate and adaptive immune cells as well as adipocytes in CrF promote fibrosis in the affected intestine by secreting large amount of pro-fibrotic cytokines, adipokines, growth factors and fatty acids. CrF is a potential therapeutic target for CD treatment and a promising bio-marker for predicting response to drug therapy. This review aims to summarize and update the clinical manifestation and application of CrF and the underlying molecular mechanism involved in the pathogenesis of intestinal inflammation and fibrosis in CD.
Collapse
Affiliation(s)
- Yi Yin
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Ying Xie
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Wei Ge
- Department of General SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsuChina
| | - Yi Li
- Department of General SurgeryJinling HospitalMedical School of Nanjing UniversityNanjingChina
| |
Collapse
|
121
|
Wang X, Jiang M, Li D, Xu L. Analyzing the Therapeutic Mechanism of Mongolian Medicine Zhonglun-5 in Rheumatoid Arthritis Using a Bagging Algorithm with Serum Metabonomics. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5997562. [PMID: 36532854 PMCID: PMC9750765 DOI: 10.1155/2022/5997562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/07/2022] [Accepted: 11/23/2022] [Indexed: 10/07/2023]
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disorder. Zhonglun-5 (ZL), a traditional Mongolian medicine, exhibits an excellent clinical effect on RA; however, its molecular mechanism remains unclear. In this study, rat serum metabolomic analysis was performed to identify potential biomarkers for RA and investigate its treatment mechanism. A Dionex Ultimate 3000 ultrahigh-performance liquid chromatography system coupled with a Q-Exactive Focus Orbitrap mass spectrometer was used for metabonomics analysis. Bootstrap aggregation (bagging) classification algorithm was applied to process data from control (CG), model (MG), and treatment administration groups. The classification accuracy was 100.00% (6/6) in the decision tree model and 83.33% (5/6) in the K-nearest neighbor (KNN) model, accompanied by 18 training samples and 6 testing samples. Using volcanic map analysis, 24 biomarkers were identified between CG and MG, including those related to glycosphingolipid biosynthesis, arachidonic acid, fatty acids, amino acids, bile acids, vitamins, and sphingolipids. A set diagram of the heatmap and drug-biomarker network of potential biomarkers was constructed. After ZL administration, the levels of these biomarkers returned to normal, indicating that ZL had a therapeutic effect in rats with RA. This study established a solid theoretical foundation to promote further research on the clinical applicability of ZL.
Collapse
Affiliation(s)
- Xiye Wang
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, China
- Inner Mongolia Key Laboratory of Chemistry for Natural Products Chemistry and Synthesis for Functional Molecules, Tongliao 028000, China
| | - Mingyang Jiang
- College of Computer Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Dan Li
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, China
- Inner Mongolia Key Laboratory of Chemistry for Natural Products Chemistry and Synthesis for Functional Molecules, Tongliao 028000, China
| | - Liang Xu
- Inner Mongolia Key Laboratory of Chemistry for Natural Products Chemistry and Synthesis for Functional Molecules, Tongliao 028000, China
| |
Collapse
|
122
|
Alsaleh G, Richter FC, Simon AK. Age-related mechanisms in the context of rheumatic disease. Nat Rev Rheumatol 2022; 18:694-710. [PMID: 36329172 DOI: 10.1038/s41584-022-00863-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
Ageing is characterized by a progressive loss of cellular function that leads to a decline in tissue homeostasis, increased vulnerability and adverse health outcomes. Important advances in ageing research have now identified a set of nine candidate hallmarks that are generally considered to contribute to the ageing process and that together determine the ageing phenotype, which is the clinical manifestation of age-related dysfunction in chronic diseases. Although most rheumatic diseases are not yet considered to be age related, available evidence increasingly emphasizes the prevalence of ageing hallmarks in these chronic diseases. On the basis of the current evidence relating to the molecular and cellular ageing pathways involved in rheumatic diseases, we propose that these diseases share a number of features that are observed in ageing, and that they can therefore be considered to be diseases of premature or accelerated ageing. Although more data are needed to clarify whether accelerated ageing drives the development of rheumatic diseases or whether it results from the chronic inflammatory environment, central components of age-related pathways are currently being targeted in clinical trials and may provide a new avenue of therapeutic intervention for patients with rheumatic diseases.
Collapse
Affiliation(s)
- Ghada Alsaleh
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK.
- Botnar Research Centre, NDORMS, University of Oxford, Oxford, UK.
| | - Felix C Richter
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Anna K Simon
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
123
|
Nanozyme-reinforced hydrogel as a H 2O 2-driven oxygenerator for enhancing prosthetic interface osseointegration in rheumatoid arthritis therapy. Nat Commun 2022; 13:6758. [PMID: 36351899 PMCID: PMC9646710 DOI: 10.1038/s41467-022-34481-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2022] Open
Abstract
Stem cell-based therapy has drawn attention for enhancing the osseointegration efficiency after joint replacement in the rheumatoid arthritis (RA). However, therapeutic efficacy of this approach is threatened by the accumulated reactive oxygen species (ROS) and poor oxygen supply. Herein, we develop a nanozyme-reinforced hydrogel for reshaping the hostile RA microenvironment and improving prosthetic interface osseointegration. The engineered hydrogel not only scavenges endogenously over-expressed ROS, but also synergistically produces dissolved oxygen. Such performance enables the hydrogel to be utilized as an injectable delivery vehicle of bone marrow-derived mesenchymal stem cells (BMSCs) to protect implanted cells from ROS and hypoxia-mediated death and osteogenic limitation. This nanozyme-reinforced hydrogel encapsulated with BMSCs can alleviate the symptoms of RA, including suppression of local inflammatory cytokines and improvement of osseointegration. This work provides a strategy for solving the long-lasting challenge of stem cell transplantation and revolutionizes conventional intervention methods for improving prosthetic interface osseointegration in RA.
Collapse
|
124
|
Li H, Yu L, Zhang X, Shang J, Duan X. Exploring the molecular mechanisms and shared gene signatures between rheumatoid arthritis and diffuse large B cell lymphoma. Front Immunol 2022; 13:1036239. [PMID: 36389761 PMCID: PMC9659608 DOI: 10.3389/fimmu.2022.1036239] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 07/25/2023] Open
Abstract
The relationship between rheumatoid arthritis (RA) and diffuse large B-cell lymphoma (DLBCL) is well characterized, but the molecular mechanisms underlying this association have not been clearly investigated. Our study aimed to identify shared gene signatures and molecular mechanisms between RA and DLBCL. We selected multiple Gene Expression Omnibus (GEO) datasets (GSE93272, GSE83632, GSE12453, GSE1919) to obtain gene expression levels and clinical information about patients with RA and DLBCL. Weighted gene co-expression network analysis (WGCNA) was used to research co-expression networks associated with RA and DLBCL. Subsequently, we performed enrichment analysis of shared genes and screened the most significant core genes. We observed expression of the screened target gene, galectin 2 (LGALS2), in DLBCL patients and its impact on patient prognosis. Finally, we analyzed the molecular functional mechanism of LGALS2 and observed its relationship with the immune response in DLBCL using single-sample Gene Set Enrichment Analysis (ssGSEA). WGCNA recognized two major modules for RA and DLBCL, respectively. Shared genes (551) were identified for RA and DLBCL by observing the intersection. In addition, a critical shared gene, LGALS2, was acquired in the validation tests. Next, we found that the expression level of LGALS2 gradually decreased with tumor progression in DLBCL and that increased expression of LGALS2 predicted a better prognosis for DLBCL patients. ssGSEA revealed that LGALS2 is involved in immune-related pathways and has a significant regulatory effect on human immune responses. Additionally, we observed that LGALS2 is closely related to the sensitivity of multiple chemotherapeutic drugs. There is extremely little research on the molecular mechanism of correlation between RA and DLBCL. Our study identified that LGALS2 is a potential therapeutic target and an immune-related biomarker for patients with RA and DLBCL.
Collapse
|
125
|
De Benedittis G, Latini A, Colafrancesco S, Priori R, Perricone C, Novelli L, Borgiani P, Ciccacci C. Alteration of Mitochondrial DNA Copy Number and Increased Expression Levels of Mitochondrial Dynamics-Related Genes in Sjögren's Syndrome. Biomedicines 2022; 10:2699. [PMID: 36359219 PMCID: PMC9687724 DOI: 10.3390/biomedicines10112699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/26/2023] Open
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune multifactorial disease characterized by inflammation and lymphocytic infiltration of the exocrine glands. Several studies have highlighted the involvement of oxidative stress in this pathology, suggesting that it could induce mitochondrial dysfunctions. Mitochondria could have a role in inflammatory and immune processes. Since the mitochondrial DNA (mtDNA) copy number could change in response to physiological or environmental stimuli, this study aimed to evaluate possible alterations in the mtDNA copy number in SS. We have analyzed the amount of mtDNA in the peripheral blood of 74 SS patients and 61 healthy controls by qPCR. Then, since mitochondrial fusion and fission play a crucial role in maintaining the number of mitochondria, we investigated the expression variability of the genes most commonly involved in mitochondrial dynamics in a subgroup of SS patients and healthy controls. Interestingly, we observed a highly significant decrease in mtDNA copies in the SS patients compared to healthy controls (p = 1.44 × 10-12). Expression levels of mitochondrial fission factor (MFF), mitofusin-1 (MFN1), and mitochondrial transcription factor A (TFAM) genes were analyzed, showing a statistically significant increase in the expression of MFF (p = 0.003) and TFAM (p = 0.022) in the SS patients compared to healthy controls. These results give further insight into the possible involvement of mitochondrial dysfunctions in SS disease.
Collapse
Affiliation(s)
- Giada De Benedittis
- Genetics Section, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Andrea Latini
- Genetics Section, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Serena Colafrancesco
- Division of Rheumatology, Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University, 00133 Rome, Italy
| | - Roberta Priori
- Division of Rheumatology, Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University, 00133 Rome, Italy
- UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Carlo Perricone
- Rheumatology Department of Medicine, University of Perugia, Piazzale Giorgio Menghini 1, 06129 Perugia, Italy
| | - Lucia Novelli
- UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Paola Borgiani
- Genetics Section, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Cinzia Ciccacci
- UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| |
Collapse
|
126
|
Lu Y, Hao C, Yu S, Ma Z, Fu X, Qin M, Ding M, Xu Z, Fan L. Cationic amino acid transporter-1 (CAT-1) promotes fibroblast-like synoviocyte proliferation and cytokine secretion by taking up L-arginine in rheumatoid arthritis. Arthritis Res Ther 2022; 24:234. [PMID: 36253807 PMCID: PMC9575222 DOI: 10.1186/s13075-022-02921-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/06/2022] [Indexed: 11/29/2022] Open
Abstract
Background
Abnormal proliferation of fibroblast-like synoviocytes (FLSs) in the synovial lining layer is the primary cause of synovial hyperplasia and joint destruction in rheumatoid arthritis (RA). Currently, the relationship between metabolic abnormalities and FLS proliferation is a new focus of investigation. However, little is known regarding the relationship between amino acid metabolism and RA. Methods The concentrations of amino acids and cytokines in the synovial fluid of RA (n = 9) and osteoarthritis (OA, n = 9) were detected by LC–MS/MS and CBA assay, respectively. The mRNA and protein expression of cationic amino acid transporter-1 (CAT-1) were determined in FLSs isolated from RA and OA patients by real-time PCR and western blotting. MTT assay, cell cycle, apoptosis, invasion, and cytokine secretion were determined in FLSs knocked down of CAT-1 using siRNA or treated with D-arginine under normoxic and hypoxic culture conditions. A mouse collagen-induced arthritis (CIA) model was applied to test the therapeutic potential of blocking the uptake of L-arginine in vivo. Results L-rginine was upregulated in the synovial fluid of RA patients and was positively correlated with the elevation of the cytokines IL-1β, IL-6, and IL-8. Further examination demonstrated that CAT-1 was the primary transporter for L-arginine and was overexpressed on RA FLSs compared to OA FLSs. Moreover, knockdown of CAT-1 using siRNA or inhibition of L-arginine uptake using D-arginine significantly suppressed L-arginine metabolism, cell proliferation, migration, and cytokine secretion in RA FLSs under normoxic and hypoxic culture conditions in vitro but increased cell apoptosis in a dose-dependent manner. Meanwhile, in vivo assays revealed that an L-arginine-free diet or blocking the uptake of L-arginine using D-arginine suppressed arthritis progression in CIA mice. Conclusion CAT-1 is upregulated and promotes FLS proliferation by taking up L-arginine, thereby promoting RA progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02921-8.
Collapse
Affiliation(s)
- Ying Lu
- Clinical Laboratory Department, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China. .,Shanghai East Hospital Ji'an Hospital, 80 Ji'an South Road, Ji'an City, 343000, Jiangxi Province, China.
| | - Chongbo Hao
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Shanshan Yu
- Clinical Laboratory Department, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Zuan Ma
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Xuelian Fu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Mingqing Qin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Menglei Ding
- Clinical Laboratory Department, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China
| | - Zengguang Xu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China.
| | - Lieying Fan
- Clinical Laboratory Department, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, China.
| |
Collapse
|
127
|
Silva Santos Ribeiro P, Willemen HLDM, Eijkelkamp N. Mitochondria and sensory processing in inflammatory and neuropathic pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1013577. [PMID: 36324872 PMCID: PMC9619239 DOI: 10.3389/fpain.2022.1013577] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023] Open
Abstract
Rheumatic diseases, such as osteoarthritis and rheumatoid arthritis, affect over 750 million people worldwide and contribute to approximately 40% of chronic pain cases. Inflammation and tissue damage contribute to pain in rheumatic diseases, but pain often persists even when inflammation/damage is resolved. Mechanisms that cause this persistent pain are still unclear. Mitochondria are essential for a myriad of cellular processes and regulate neuronal functions. Mitochondrial dysfunction has been implicated in multiple neurological disorders, but its role in sensory processing and pain in rheumatic diseases is relatively unexplored. This review provides a comprehensive understanding of how mitochondrial dysfunction connects inflammation and damage-associated pathways to neuronal sensitization and persistent pain. To provide an overall framework on how mitochondria control pain, we explored recent evidence in inflammatory and neuropathic pain conditions. Mitochondria have intrinsic quality control mechanisms to prevent functional deficits and cellular damage. We will discuss the link between neuronal activity, mitochondrial dysfunction and chronic pain. Lastly, pharmacological strategies aimed at reestablishing mitochondrial functions or boosting mitochondrial dynamics as therapeutic interventions for chronic pain are discussed. The evidence presented in this review shows that mitochondria dysfunction may play a role in rheumatic pain. The dysfunction is not restricted to neuronal cells in the peripheral and central nervous system, but also includes blood cells and cells at the joint level that may affect pain pathways indirectly. Pre-clinical and clinical data suggest that modulation of mitochondrial functions can be used to attenuate or eliminate pain, which could be beneficial for multiple rheumatic diseases.
Collapse
Affiliation(s)
| | | | - Niels Eijkelkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
128
|
Jo Y, Woo JS, Lee AR, Lee SY, Shin Y, Lee LP, Cho ML, Kang T. Inner-Membrane-Bound Gold Nanoparticles as Efficient Electron Transfer Mediators for Enhanced Mitochondrial Electron Transport Chain Activity. NANO LETTERS 2022; 22:7927-7935. [PMID: 36137175 DOI: 10.1021/acs.nanolett.2c02957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Electron transfer through the mitochondrial electron transport chain (ETC) can be critically blocked by the dysfunction of protein complexes. Redox-active molecules have been used to mediate the electron transfer in place of the dysfunctional complexes; however, they are limited to replacing complex I and are known to be toxic. Here we report artificial mitochondrial electron transfer pathways that enhance ETC activity by exploiting inner-membrane-bound gold nanoparticles (GNPs) as efficient electron transfer mediators. The hybridization of mitochondria with GNPs, driven by electrostatic interaction, is successfully visualized in real time at the level of a single mitochondrion. By observing quantized quenching dips via plasmon resonance energy transfer, we reveal that the hybridized GNPs are bound to the inner membrane of mitochondria irrespective of the presence of the outer membrane. The ETC activity of mitochondria with GNPs such as membrane potential, oxygen consumption, and ATP production is remarkably increased in vitro.
Collapse
Affiliation(s)
- Yuseung Jo
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea
| | - Jin Seok Woo
- Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea
| | - A Ram Lee
- Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seon-Yeong Lee
- Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea
| | - Yonghee Shin
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea
| | - Luke P Lee
- Harvard Medical School, Harvard University; Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Department of Bioengineering, and Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, California 94720, United States
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Mi-La Cho
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Medical Life Scieneces, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Taewook Kang
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| |
Collapse
|
129
|
Zhao H, Li L, Zhao N, Lu A, Lu C, He X. The effect of long non-coding RNAs in joint destruction of rheumatoid arthritis. Front Cell Dev Biol 2022; 10:1011371. [PMID: 36263019 PMCID: PMC9574091 DOI: 10.3389/fcell.2022.1011371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 11/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease accompanied with joint destruction. Serious joint destruction will eventually lead to disability and the decline of life quality in RA patients. At present, the therapeutic effect of drugs to alleviate joint destruction in RA is limited. Recently, accumulating evidences have shown that long non-coding RNAs (lncRNAs) play an important role in the pathogenesis of joint diseases. Therefore, this paper reviews the expression change and the action mechanism of lncRNAs in joint destruction of RA in recent years. A more comprehensive understanding of the role of lncRNAs in joint destruction will help the treatment of RA.
Collapse
Affiliation(s)
- Hanxiao Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
- *Correspondence: Aiping Lu, ; Cheng Lu, ; Xiaojuan He,
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Aiping Lu, ; Cheng Lu, ; Xiaojuan He,
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Aiping Lu, ; Cheng Lu, ; Xiaojuan He,
| |
Collapse
|
130
|
Deng M, Wu Y, Ren Y, Song H, Zheng L, Lin G, Wen X, Tao Y, Kong Q, Wang Y. Clickable and smart drug delivery vehicles accelerate the healing of infected diabetic wounds. J Control Release 2022; 350:613-629. [PMID: 36058354 DOI: 10.1016/j.jconrel.2022.08.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 11/15/2022]
Abstract
In this study, an adipic acid dihydrazide (ADH)/ tannic acid (TA)-grafted hyaluronic acid (HA)-based multifunctional hydrogel was synthesized through a spontaneous amino-yne click reaction and used to promote the improved healing of infected diabetic wounds. This hydrogel exhibited a range of beneficial properties such as tunable gelation time, adjustable mechanical properties, pH-sensitive response characteristics, excellent injectability, the ability to readily adhere to tissue, and ultra-intimate contact capabilities. Following the encapsulation of ultrasmall Ag nanoclusters (AgNCs) and deferoxamine loaded polydopamine/ hollow mesoporous manganese dioxide (PHMD, PDA/H-mMnO2@DFO) nanoparticles, the prepared hydrogel presented with robust antibacterial, anti-inflammatory, and pro-angiogenic properties and a desirable smart drug release profile. In this fabricated platform, PHMD was able to effectively alleviate localized oxidative stress and prolonged oxygen deprivation via the decomposition of endogenous H2O2 to produce O2. Further in vivo assays revealed that this hydrogel was capable of facilitating the healing of infected wounds through the sequential engagement of antibacterial, anti-inflammatory, and pro-angiogenic activities. Together, this synthesized clickable environmentally-responsive hydrogel offers great promise as a tool that can be applied to aid in the healing of chronically infected diabetic wounds and other inflammatory conditions.
Collapse
Affiliation(s)
- Mingyan Deng
- WestChina-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ye Wu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Ren
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haoyang Song
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Zheng
- WestChina-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guangzhi Lin
- WestChina-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Wen
- WestChina-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiran Tao
- WestChina-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingquan Kong
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
131
|
Manda G, Milanesi E, Genc S, Niculite CM, Neagoe IV, Tastan B, Dragnea EM, Cuadrado A. Pros and cons of NRF2 activation as adjunctive therapy in rheumatoid arthritis. Free Radic Biol Med 2022; 190:179-201. [PMID: 35964840 DOI: 10.1016/j.freeradbiomed.2022.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with an important inflammatory component accompanied by deregulated redox-dependent signaling pathways that are feeding back into inflammation. In this context, we bring into focus the transcription factor NRF2, a master redox regulator that exerts exquisite antioxidant and anti-inflammatory effects. The review does not intend to be exhaustive, but to point out arguments sustaining the rationale for applying an NRF2-directed co-treatment in RA as well as its potential limitations. The involvement of NRF2 in RA is emphasized through an analysis of publicly available transcriptomic data on NRF2 target genes and the findings from NRF2-knockout mice. The impact of NRF2 on concurrent pathologic mechanisms in RA is explained by its crosstalk with major redox-sensitive inflammatory and cell death-related pathways, in the context of the increased survival of pathologic cells in RA. The proposed adjunctive therapy targeted to NRF2 is further sustained by the existence of promising NRF2 activators that are in various stages of drug development. The interference of NRF2 with conventional anti-rheumatic therapies is discussed, including the cytoprotective effects of NRF2 for alleviating drug toxicity. From another perspective, the review presents how NRF2 activation would be decreasing the efficacy of synthetic anti-rheumatic drugs by increasing drug efflux. Future perspectives regarding pharmacologic NRF2 activation in RA are finally proposed.
Collapse
Affiliation(s)
- Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Elena Milanesi
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Sermin Genc
- Neurodegeneration and Neuroprotection Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey; Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Izmir, Turkey
| | - Cristina Mariana Niculite
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania; Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Ionela Victoria Neagoe
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Bora Tastan
- Neurodegeneration and Neuroprotection Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Elena Mihaela Dragnea
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
132
|
Ma C, Wang J, Hong F, Yang S. Mitochondrial Dysfunction in Rheumatoid Arthritis. Biomolecules 2022; 12:biom12091216. [PMID: 36139055 PMCID: PMC9496243 DOI: 10.3390/biom12091216] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/14/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis, a chronic autoimmune disease with complex etiology, is characterized by excessive proliferation of synovial cells, massive production of inflammatory cells and cartilage destruction. Studies have shown that mitochondrial dysfunction plays an important role in promoting the occurrence of RA. Mitochondria with normal structure and function are essential for the normal survival of chondrocytes and synovial cells. Once mitochondrial function is destroyed, it will affect the survival, activation and differentiation of immune cells and non-immune cells involved in the pathogenesis of RA, thus leading to the occurrence of RA. However, the mechanism of mitochondrial dysfunction in RA remains unclear. This article reviews the method of mitochondrial dysfunction leading to RA, the effects of mitochondrial dysfunction on immune cells, the etiology of mitochondrial dysfunction in RA, and the pathology of mitochondrial dysfunction in RA. We also outline some drugs that can exert therapeutic effects on RA which are associated with modulating mitochondrial activity. The understanding and summary of mitochondrial dysfunction in RA may provide new research directions for pathological intervention and prevention of RA.
Collapse
Affiliation(s)
- Chen Ma
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China
- Queen Mary School, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Jie Wang
- Department of Graduate, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Fenfang Hong
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China
- Correspondence: (F.H.); or (S.Y.)
| | - Shulong Yang
- Key Laboratory of Chronic Diseases, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Correspondence: (F.H.); or (S.Y.)
| |
Collapse
|
133
|
Citrullination: A modification important in the pathogenesis of autoimmune diseases. Clin Immunol 2022; 245:109134. [DOI: 10.1016/j.clim.2022.109134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
|
134
|
Icariin represses the inflammatory responses and survival of rheumatoid arthritis fibroblast-like synoviocytes by regulating the TRIB1/TLR2/NF-kB pathway. Int Immunopharmacol 2022; 110:108991. [DOI: 10.1016/j.intimp.2022.108991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022]
|
135
|
Zhou J, Mao Y, Shi X, Zhang Y, Yu X, Liu X, Diao L, Yang X, Liu C, Liu D, Tan X, Liu M. Peimine suppresses collagen-induced arthritis, activated fibroblast-like synoviocytes and TNFα-induced MAPK pathways. Int Immunopharmacol 2022; 111:109181. [PMID: 36027853 DOI: 10.1016/j.intimp.2022.109181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND PURPOSE Peimine (PM), a main isosterol alkaloid component isolated from the bulbs of traditional Chinese herb Fritillaria cirrhosa D. Don, has been demonstrated to exhibit multiple pharmacological properties, including anti-inflammation, anti-cancer and pain suppression. However, its effect on rheumatoid arthritis (RA) remains unknown. In the present study, we investigated the effect of PM on collagen-induced arthritis (CIA) rats in vivo and its inhibition on destructive behaviors of arthritic fibroblast-like synoviocytes (FLSs) in vitro. METHODS Arthritis was induced in rats by chicken type II collagen. Arthritis score, radiological evaluation, and histopathological assessment were used to evaluate the therapeutic effects of PM on CIA rats. EdU assay, wound healing assay and real-time PCR were used to examine the inhibitory effect of PM on proliferation, migration, and over-expression of pro-inflammatory cytokines in TNFα-induced arthritic FLSs. TRAP staining and scanning electron microscopy were used to analyze the effect of PM on osteoclastogensis and bone resorption. Western blot was used to reveal PM's molecular mechanism of action on RA. RESULTS PM significantly suppressed synovitis and bone destruction in CIA rats. In vitro experiments showed that PM treatment significantly inhibited TNFα-induced destructive behaviors of arthritic FLSs, including over-proliferation, migration and over-expression of pro-inflammatory cytokines. Additionally, RANKL-induced osteoclast formation and bone-resorpting function were also inhibited by PM. Further molecular mechanism studies revealed that PM treatment significantly suppressed TNFα-induced activations of MAPKs (ERK, JNK and p38) in arthritic FLSs. CONCLUSION Our findings provide strong evidence that PM has the potential to be developed as a therapeutic agent for patients with RA.
Collapse
Affiliation(s)
- Junnan Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yuhang Mao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xiaotian Shi
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yudie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xiaolu Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xuan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Li Diao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xue Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Changze Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Dan Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Xin Tan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China
| | - Mei Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
136
|
Li S, Ma R, Hu XY, Li HB, Geng WC, Kong X, Zhang C, Guo DS. Drug in Drug: A Host-Guest Formulation of Azocalixarene with Hydroxychloroquine for Synergistic Anti-Inflammation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2203765. [PMID: 35680644 DOI: 10.1002/adma.202203765] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Macrocyclic delivery and therapeutics are two significant topics in supramolecular biomedicine. The functional integration of these topics would open new avenues for treating diseases synergistically. However, these two individual topics have only been occasionally merged, probably because of the lack of functionalized design of macrocyclic host and the lack of efficient recognition between host and guest drugs. Herein, a "drug-in-drug" strategy is proposed, in which an active drug is encapsulated by a macrocycle possessing therapeutic activity to form a multifunctional supramolecular active pharmaceutical ingredient. As a proof-of-concept, a complex of hydroxychloroquine (HCQ) with sulfonated azocalix[4]arene (HCQ@SAC4A) is prepared to treat rheumatoid arthritis (RA) in a combined fashion. SAC4A is a therapeutic agent that exhibits scavenging capacity for reactive oxygen species and exerts an anti-inflammatory effect. It is also a hypoxia-responsive carrier that can deliver HCQ directly to the inflammatory articular cavity. Consequently, HCQ@SAC4A achieves the synergistic anti-inflammatory effect on both inflamed RAW 264.7 cells and RA rats. This effect is attributed to the temporal and spatial consistency of the two active ingredients of the complex. As a new paradigm for combinational therapy, the drug-in-drug strategy advances in easy preparation, mix-and-match combination, and precise ratiometric control.
Collapse
Affiliation(s)
- Shihui Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Rong Ma
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Hua-Bin Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Wen-Chao Geng
- Key Laboratory of Systems Microbial Biotechnology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
| | - Xianglei Kong
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Chao Zhang
- Orthopedics Department, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300380, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
137
|
Li Y, Liang Q, Zhou L, Cao Y, Yang J, Li J, Liu J, Bi J, Liu Y. An ROS-responsive artesunate prodrug nanosystem co-delivers dexamethasone for rheumatoid arthritis treatment through the HIF-1α/NF-κB cascade regulation of ROS scavenging and macrophage repolarization. Acta Biomater 2022; 152:406-424. [DOI: 10.1016/j.actbio.2022.08.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/17/2022] [Accepted: 08/23/2022] [Indexed: 11/01/2022]
|
138
|
Chang S, Tang M, Zhang B, Xiang D, Li F. Ferroptosis in inflammatory arthritis: A promising future. Front Immunol 2022; 13:955069. [PMID: 35958605 PMCID: PMC9361863 DOI: 10.3389/fimmu.2022.955069] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/04/2022] [Indexed: 12/13/2022] Open
Abstract
Ferroptosis is a kind of regulatory cell death (RCD) caused by iron accumulation and lipid peroxidation, which is characterized by mitochondrial morphological changes and has a complex regulatory network. Ferroptosis has been gradually emphasized in the pathogenesis of inflammatory arthritis. In this review, we summarized the relevant research on ferroptosis in various inflammatory arthritis including rheumatoid arthritis (RA), osteoarthritis, gout arthritis, and ankylosing spondylitis, and focused on the relationship between RA and ferroptosis. In patients with RA and animal models of RA, there was evidence of iron overload and lipid peroxidation, as well as mitochondrial dysfunction that may be associated with ferroptosis. Ferroptosis inducers have shown good application prospects in tumor therapy, and some anti-rheumatic drugs such as methotrexate and sulfasalazine have been shown to have ferroptosis modulating effects. These phenomena suggest that the role of ferroptosis in the pathogenesis of inflammatory arthritis will be worth further study. The development of therapeutic strategies targeting ferroptosis for patients with inflammatory arthritis may be a promising future.
Collapse
Affiliation(s)
- Siyuan Chang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mengshi Tang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fen Li
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Fen Li,
| |
Collapse
|
139
|
Blagov AV, Grechko AV, Nikiforov NG, Zhuravlev AD, Sadykhov NK, Orekhov AN. Effects of Metabolic Disorders in Immune Cells and Synoviocytes on the Development of Rheumatoid Arthritis. Metabolites 2022; 12:metabo12070634. [PMID: 35888759 PMCID: PMC9324614 DOI: 10.3390/metabo12070634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/30/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease that affects the joints. It has been proven that, with the development of RA, there are changes in the metabolism of cells located in the focus of inflammation. In this article, we describe the connection between metabolism and inflammation in the context of rheumatoid arthritis. We consider in detail the changes in metabolic processes and their subsequent immunomodulatory effects. In particular, we consider how changes in mitochondrial functioning lead to the modulation of metabolism in rheumatoid arthritis. We also describe the main features of the metabolism in cells present in the synovial membrane during inflammation, and we discuss possible targets for the therapy of rheumatoid arthritis.
Collapse
Affiliation(s)
- Alexander V. Blagov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
- Correspondence: (A.V.B.); (A.N.O.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14–3 Solyanka Street, 109240 Moscow, Russia;
| | - Nikita G. Nikiforov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
| | - Alexander D. Zhuravlev
- Petrovsky National Research Centre of Surgery, AP Avtsyn Institute of Human Morphology, 117418 Moscow, Russia;
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
- Correspondence: (A.V.B.); (A.N.O.)
| |
Collapse
|
140
|
Jávor P, Mácsai A, Butt E, Baráth B, Jász DK, Horváth T, Baráth B, Csonka Á, Török L, Varga E, Hartmann P. Mitochondrial Dysfunction Affects the Synovium of Patients with Rheumatoid Arthritis and Osteoarthritis Differently. Int J Mol Sci 2022; 23:ijms23147553. [PMID: 35886901 PMCID: PMC9319158 DOI: 10.3390/ijms23147553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
There is growing evidence regarding the role of mitochondrial dysfunction in osteoarthritis (OA) and rheumatoid arthritis (RA). However, quantitative comparison of synovial mitochondrial derangements in these main arthritis forms is missing. A prospective clinical study was conducted on adult patients undergoing knee surgery. Patients were allocated into RA and OA groups based on disease-specific clinical scores, while patients without arthritis served as controls. Synovial samples were subjected to high-resolution respirometry to analyze mitochondrial functions. From the total of 814 patients, 109 cases were enrolled into the study (24 RA, 47 OA, and 38 control patients) between 1 September 2019 and 31 December 2021. The decrease in complex I-linked respiration and dyscoupling of mitochondria were characteristics of RA patients, while both arthritis groups displayed reduced OxPhos activity compared to the control group. However, no significant difference was found in complex II-related activity between the OA and RA groups. The cytochrome C release and H2O2 formation were increased in both arthritis groups. Mitochondrial dysfunction was present in both arthritis groups; however, to a different extent. Consequently, mitochondrial protective agents may have major benefits for arthritis patients. Based on our current study, we recommend focusing on respiratory complex I in rheumatoid arthritis research.
Collapse
Affiliation(s)
- Péter Jávor
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
| | - Attila Mácsai
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
| | - Edina Butt
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
| | - Bálint Baráth
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
- Institute of Surgical Research, University of Szeged, 6720 Szeged, Hungary; (D.K.J.); (T.H.)
| | - Dávid Kurszán Jász
- Institute of Surgical Research, University of Szeged, 6720 Szeged, Hungary; (D.K.J.); (T.H.)
| | - Tamara Horváth
- Institute of Surgical Research, University of Szeged, 6720 Szeged, Hungary; (D.K.J.); (T.H.)
| | - Bence Baráth
- Department of Pathology, University of Szeged, 6720 Szeged, Hungary;
| | - Ákos Csonka
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
| | - László Török
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
- Department of Sports Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Endre Varga
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
| | - Petra Hartmann
- Department of Traumatology, University of Szeged, 6720 Szeged, Hungary; (P.J.); (A.M.); (E.B.); (B.B.); (Á.C.); (L.T.); (E.V.)
- Correspondence:
| |
Collapse
|
141
|
Zhou X, Mi J, Liu Z. Causal association of diet-derived circulating antioxidants with the risk of rheumatoid arthritis: a Mendelian randomization study. Semin Arthritis Rheum 2022; 56:152079. [DOI: 10.1016/j.semarthrit.2022.152079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/16/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022]
|
142
|
Wang X, Fan D, Cao X, Ye Q, Wang Q, Zhang M, Xiao C. The Role of Reactive Oxygen Species in the Rheumatoid Arthritis-Associated Synovial Microenvironment. Antioxidants (Basel) 2022; 11:antiox11061153. [PMID: 35740050 PMCID: PMC9220354 DOI: 10.3390/antiox11061153] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease that begins with a loss of tolerance to modified self-antigens and immune system abnormalities, eventually leading to synovitis and bone and cartilage degradation. Reactive oxygen species (ROS) are commonly used as destructive or modifying agents of cellular components or they act as signaling molecules in the immune system. During the development of RA, a hypoxic and inflammatory situation in the synovium maintains ROS generation, which can be sustained by increased DNA damage and malfunctioning mitochondria in a feedback loop. Oxidative stress caused by abundant ROS production has also been shown to be associated with synovitis in RA. The goal of this review is to examine the functions of ROS and related molecular mechanisms in diverse cells in the synovial microenvironment of RA. The strategies relying on regulating ROS to treat RA are also reviewed.
Collapse
Affiliation(s)
- Xing Wang
- School of Clinical Medicine, China-Japan Friendship Hospital, Beijing University of Chinese Medicine, Beijing 100029, China; (X.W.); (Q.Y.); (Q.W.)
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
| | - Danping Fan
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Xiaoxue Cao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Qinbin Ye
- School of Clinical Medicine, China-Japan Friendship Hospital, Beijing University of Chinese Medicine, Beijing 100029, China; (X.W.); (Q.Y.); (Q.W.)
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
| | - Qiong Wang
- School of Clinical Medicine, China-Japan Friendship Hospital, Beijing University of Chinese Medicine, Beijing 100029, China; (X.W.); (Q.Y.); (Q.W.)
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
| | - Mengxiao Zhang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China; (D.F.); (X.C.); (M.Z.)
- Department of Emergency, China-Japan Friendship Hospital, Beijing 100029, China
- Correspondence: or
| |
Collapse
|
143
|
López-Armada MJ, Fernández-Rodríguez JA, Blanco FJ. Mitochondrial Dysfunction and Oxidative Stress in Rheumatoid Arthritis. Antioxidants (Basel) 2022; 11:antiox11061151. [PMID: 35740048 PMCID: PMC9220001 DOI: 10.3390/antiox11061151] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
Control of excessive mitochondrial oxidative stress could provide new targets for both preventive and therapeutic interventions in the treatment of chronic inflammation or any pathology that develops under an inflammatory scenario, such as rheumatoid arthritis (RA). Increasing evidence has demonstrated the role of mitochondrial alterations in autoimmune diseases mainly due to the interplay between metabolism and innate immunity, but also in the modulation of inflammatory response of resident cells, such as synoviocytes. Thus, mitochondrial dysfunction derived from several danger signals could activate tricarboxylic acid (TCA) disruption, thereby favoring a vicious cycle of oxidative/mitochondrial stress. Mitochondrial dysfunction can act through modulating innate immunity via redox-sensitive inflammatory pathways or direct activation of the inflammasome. Besides, mitochondria also have a central role in regulating cell death, which is deeply altered in RA. Additionally, multiple evidence suggests that pathological processes in RA can be shaped by epigenetic mechanisms and that in turn, mitochondria are involved in epigenetic regulation. Finally, we will discuss about the involvement of some dietary components in the onset and progression of RA.
Collapse
Affiliation(s)
- María José López-Armada
- Grupo de Investigación en Envejecimiento e Inflamación (ENVEINF), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain;
- Correspondence: (M.J.L.-A.); (F.J.B.); Tel./Fax: +34-981-178272-73 (M.J.L.-A.)
| | - Jennifer Adriana Fernández-Rodríguez
- Grupo de Investigación en Envejecimiento e Inflamación (ENVEINF), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain;
| | - Francisco Javier Blanco
- Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación de Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña, 15001 A Coruña, Spain
- Correspondence: (M.J.L.-A.); (F.J.B.); Tel./Fax: +34-981-178272-73 (M.J.L.-A.)
| |
Collapse
|
144
|
Chen D, Guo J, Li L. Catalpol promotes mitochondrial biogenesis in chondrocytes. Arch Physiol Biochem 2022; 128:802-808. [PMID: 32096418 DOI: 10.1080/13813455.2020.1727927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The chondrocyte mitochondrial dysfunction has been considered to be associated with the pathogenesis of joint diseases. Catalpol is an active traditional Chinese medicine ingredient named Di-Huang, which is used widely to treat different diseases. In this study, we found the addition of catalpol in chondrocytes induced the expression of crucial mitochondrial regulators, peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), nuclear respiratory factor-1 (NRF1), and mitochondrial transcription factor A (TFAM). Catalpol promoted mitochondrial biogenesis, as revealed by the induction on the mitochondrial DNA/nuclear DNA (mtDNA/nDNA) and the expression of several mitochondrial genes including translocase of outer mitochondrial membrane 22 (Tomm22), translocase of outer mitochondrial membrane 70 (Tomm70), mitochondrial import inner membrane translocase subunit 50 (Timm50), NADH dehydrogenase [ubiquinone] iron-sulphur protein 3 (NDUFS3), adenosine triphosphate (ATP) synthase subunit D (ATP5d), and cytochrome B. Consequently, catalpol increased cytochrome c oxidase activity, the mitochondrial respiratory rate, and the extracellular ATP production, indicating that catalpol boosted mitochondrial function. Mechanistically, catalpol increased the activation of the cAMP-responsive element-binding protein (CREB), and the inhibition of CREB abolished catalpol-mediated promotion on mitochondrial biogenesis. In summary, this study demonstrated that catalpol has the potential to be used in the treatment of joint diseases.
Collapse
Affiliation(s)
- Dan Chen
- Department of Rehabilitation, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jing Guo
- Department of Rehabilitation, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Longguang Li
- Department of Rehabilitation, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
145
|
Jiang J, Zhan X, Qu H, Liang T, Li H, Chen L, Huang S, Sun X, Jiang W, Chen J, Chen T, Yao Y, Wu S, Zhu J, Liu C. Upregulated of ANXA3, SORL1, and Neutrophils May Be Key Factors in the Progressionof Ankylosing Spondylitis. Front Immunol 2022; 13:861459. [PMID: 35464477 PMCID: PMC9019158 DOI: 10.3389/fimmu.2022.861459] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/18/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction The specific pathogenesis of ankylosing spondylitis (AS) remains unclear, and our study aimed to investigate the possible pathogenesis of AS. Materials and Methods Two datasets were downloaded from the GEO database to perform differentially expressed gene analysis, GO enrichment analysis, KEGG pathway analysis, DO enrichment analysis, GSEA analysis of differentially expressed genes, and construction of diagnostic genes using SVM and WGCNA along with Hypoxia-related genes. Also, drug sensitivity analysis was performed on diagnostic genes. To identify the differentially expressed immune genes in the AS and control groups, we analyzed the composition of immune cells between them. Then, we examined differentially expressed genes in three AS interspinous ligament specimens and three Degenerative lumbar spine specimens using high-throughput sequencing while the immune cells were examined using the neutrophil count data from routine blood tests of 1770 HLA-B27-positive samples and 7939 HLA-B27-negative samples. To assess the relationship between ANXA3 and SORL1 and disease activity, we took the neutrophil counts of the first 50 patients with above-average BASDAI scores and the last 50 patients with below-average BASDAI scores for statistical analysis. We used immunohistochemistry to verify the expression of ANXA3 and SORL1 in AS and in controls. Results ANXA3 and SORL1 were identified as new diagnostic genes for AS. These two genes showed a significant differential expression between AS and controls, along with showing a significant positive correlation with the neutrophil count. The results of high-throughput sequencing verified that these two gene deletions were indeed differentially expressed in AS versus controls. Data from a total of 9707 routine blood tests showed that the neutrophil count was significantly higher in AS patients than in controls (p < 0.001). Patients with AS with a high BASDAI score had a much higher neutrophil count than those with a low score, and the difference was statistically significant (p < 0.001). The results of immunohistochemistry showed that the expression of ANXA3 and SORL1 in AS was significantly higher than that in the control group. Conclusion Upregulated of ANXA3, SORL1, and neutrophils may be a key factor in the progression of Ankylosing spondylitis.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinli Zhan
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haishun Qu
- Department of Traditional Chinese Medicine, The People's Hospital of Guangxi Zhuang Autonmous Region, Nanning, China
| | - Tuo Liang
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hao Li
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liyi Chen
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shengsheng Huang
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xuhua Sun
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenyong Jiang
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiarui Chen
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tianyou Chen
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuanlin Yao
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shaofeng Wu
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jichong Zhu
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chong Liu
- Department of Spinal Orthopedic Surgery, The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
146
|
Fearon U, Hanlon MM, Floudas A, Veale DJ. Cellular metabolic adaptations in rheumatoid arthritis and their therapeutic implications. Nat Rev Rheumatol 2022; 18:398-414. [PMID: 35440762 DOI: 10.1038/s41584-022-00771-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Activation of endothelium and immune cells is fundamental to the initiation of autoimmune diseases such as rheumatoid arthritis (RA), and it results in trans-endothelial cell migration and synovial fibroblast proliferation, leading to joint destruction. In RA, the synovial microvasculature is highly dysregulated, resulting in inefficient oxygen perfusion to the synovium, which, along with the high metabolic demands of activated immune and stromal cells, leads to a profoundly hypoxic microenvironment. In inflamed joints, infiltrating immune cells and synovial resident cells have great requirements for energy and nutrients, and they adapt their metabolic profiles to generate sufficient energy to support their highly activated inflammatory states. This shift in metabolic capacity of synovial cells enables them to produce the essential building blocks to support their proliferation, activation and invasiveness. Furthermore, it results in the accumulation of metabolic intermediates and alteration of redox-sensitive pathways, affecting signalling pathways that further potentiate the inflammatory response. Importantly, the inflamed synovium is a multicellular tissue, with cells differing in their metabolic requirements depending on complex cell-cell interactions, nutrient supply, metabolic intermediates and transcriptional regulation. Therefore, understanding the complex interplay between metabolic and inflammatory pathways in synovial cells in RA will provide insight into the underlying mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland. .,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland.
| | - Megan M Hanlon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
147
|
Yang B, Yao H, Yang J, Chen C, Shi J. Construction of a two-dimensional artificial antioxidase for nanocatalytic rheumatoid arthritis treatment. Nat Commun 2022; 13:1988. [PMID: 35418125 PMCID: PMC9008001 DOI: 10.1038/s41467-022-29735-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/28/2022] [Indexed: 12/15/2022] Open
Abstract
Constructing nanomaterials mimicking the coordination environments of natural enzymes may achieve biomimetic catalysis. Here we construct a two-dimensional (2D) metal-organic framework (MOF) nanosheet catalyst as an artificial antioxidase for nanocatalytic rheumatoid arthritis treatment. The 2D MOF periodically assembles numbers of manganese porphyrin molecules, which has a metal coordination geometry analogous to those of two typical antioxidases, human mitochondrial manganese superoxide dismutase (Mn-SOD) and human erythrocyte catalase. The zinc atoms of the 2D MOF regulate the metal-centered redox potential of coordinated manganese porphyrin ligand, endowing the nanosheet with both SOD- and catalase-like activities. Cellular experiments show unique anti-inflammatory and pro-biomineralization performances of the 2D MOF, while in vivo animal model further demonstrates its desirable antiarthritic efficacy. It is expected that such a nanocatalytic antioxidation concept may provide feasible approaches to future anti-inflammatory treatments.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Heliang Yao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China
| | - Jiacai Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chang Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China.
- Tenth People's Hospital and Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200092, P. R. China.
| |
Collapse
|
148
|
Wo L, Zhang X, Ma C, Zhou C, Li J, Hu Z, Gong X, Zhan M, He M, Zhao Q. LncRNA HABON promoted liver cancer cells survival under hypoxia by inhibiting mPTP opening. Cell Death Dis 2022; 8:171. [PMID: 35387966 PMCID: PMC8986810 DOI: 10.1038/s41420-022-00917-6] [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: 12/07/2021] [Revised: 02/11/2022] [Accepted: 02/24/2022] [Indexed: 12/11/2022]
Abstract
Hypoxia is an important feature of the tumor microenvironment (TME). While targeting hypoxic TME is emerging as a potential strategy for treating solid tumors including liver cancer. Recent studies have shown that hypoxia can regulate tumor adaptation to hypoxic TME through long non-coding RNA (lncRNA). In the previous study, we identify a novel hypoxia-activated lncRNA and termed it as HABON. Here, we demonstrated that knockdown of HABON caused necroptosis of tumor tissue and inhibited the subcutaneous tumor growth of SMMC-7721 cells in nude mice. Moreover, knockdown of HABON increased RIPK1 and MLKL expression as well as their phosphorylation level in SMMC-7721 and Huh7 liver cancer cells. Meanwhile, Necrostatin-1 and GSK872 could restore cell death of liver cancer cells caused by knockdown of HABON under hypoxia. The above results suggested that HABON could inhibit hypoxia-induced necroptosis of liver cancer cells. Mechanically, knockdown of HABON in liver cancer cells aggravated mitochondrial dysfunction caused by hypoxia. Furthermore, the RNA pull-down combined with mass spectrometry analysis identified HABON can interact with mitochondria-related protein VDAC1 and the RNA immunoprecipitation (RIP) analysis proved the interaction. In addition, we proved that VDAC1 mediated the mitochondrial permeability transition pore (mPTP) opening, mitochondrial dysfunction, as well as necroptosis caused by knockdown of HABON. Overall, our work demonstrates HABON can reduce hypoxia-induced necroptosis of liver cancer cells and suggests that inhibition of HABON in the hypoxic TME is a potential therapeutic strategy for treating liver cancer.
Collapse
Affiliation(s)
- Lulu Wo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Xin Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Chengning Ma
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Cixiang Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Jingchi Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Zhexuan Hu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Xiufeng Gong
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China
| | - Mengna Zhan
- Department of Pathology, Zhongshan Hospital, Fudan University, 200031, Shanghai, China
| | - Ming He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China.
| | - Qian Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Diseases, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 200025, Shanghai, China.
| |
Collapse
|
149
|
Floudas A, Gorman A, Neto N, Monaghan MG, Elliott Z, Fearon U, Marzaioli V. Inside the Joint of Inflammatory Arthritis Patients: Handling and Processing of Synovial Tissue Biopsies for High Throughput Analysis. Front Med (Lausanne) 2022; 9:830998. [PMID: 35372383 PMCID: PMC8967180 DOI: 10.3389/fmed.2022.830998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/04/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory arthritis is a chronic systemic autoimmune disease of unknown etiology, which affects the joints. If untreated, these diseases can have a detrimental effect on the patient's quality of life, leading to disabilities, and therefore, exhibit a significant socioeconomic impact and burden. While studies of immune cell populations in arthritis patient's peripheral blood have been informative regarding potential immune cell dysfunction and possible patient stratification, there are considerable limitations in identifying the early events that lead to synovial inflammation. The joint, as the site of inflammation and the local microenvironment, exhibit unique characteristics that contribute to disease pathogenesis. Understanding the contribution of immune and stromal cell interactions within the inflamed joint has been met with several technical challenges. Additionally, the limited availability of synovial tissue biopsies is a key incentive for the utilization of high-throughput techniques in order to maximize information gain. This review aims to provide an overview of key methods and novel techniques that are used in the handling, processing and analysis of synovial tissue biopsies and the potential synergy between these techniques. Herein, we describe the utilization of high dimensionality flow cytometric analysis, single cell RNA sequencing, ex vivo functional assays and non-intrusive metabolic characterization of synovial cells on a single cell level based on fluorescent lifetime imaging microscopy. Additionally, we recommend important points of consideration regarding the effect of different storage and handling techniques on downstream analysis of synovial tissue samples. The introduction of new powerful techniques in the study of synovial tissue inflammation, brings new challenges but importantly, significant opportunities. Implementation of novel approaches will accelerate our path toward understanding of the mechanisms involved in the pathogenesis of inflammatory arthritis and lead to the identification of new avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Achilleas Floudas
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- European League Against Rheumatism (EULAR) Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin (UCD), Dublin, Ireland
- *Correspondence: Achilleas Floudas
| | - Aine Gorman
- European League Against Rheumatism (EULAR) Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin (UCD), Dublin, Ireland
| | - Nuno Neto
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Michael G. Monaghan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Zoe Elliott
- European League Against Rheumatism (EULAR) Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin (UCD), Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- European League Against Rheumatism (EULAR) Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin (UCD), Dublin, Ireland
| | - Viviana Marzaioli
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- European League Against Rheumatism (EULAR) Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
150
|
Qian K, Zheng XX, Wang C, Huang WG, Liu XB, Xu SD, Liu DK, Liu MY, Lin CS. β-Sitosterol Inhibits Rheumatoid Synovial Angiogenesis Through Suppressing VEGF Signaling Pathway. Front Pharmacol 2022; 12:816477. [PMID: 35295740 PMCID: PMC8918576 DOI: 10.3389/fphar.2021.816477] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Rheumatoid arthritis (RA) is a chronic disabling inflammatory disease that causes synovial angiogenesis in an invasive manner and leads to joint destruction. Currently available pharmacotherapy for RA has unwanted side effects and limitations. Although anti-angiogenic therapy is regarded as a new potential treatment for RA, only a few anti-angiogenic drugs are available. An increasing number of studies have shown that β-sitosterol (BSS) may exert inhibitory effects against angiogenesis. However, the mechanisms involved are still unclear.Methods: Based on the results of the gene set enrichment analysis (GSEA) of the transcriptome data of endothelial cells from RA patients, we evaluated the pharmacological effects of BSS on the tube formation, cell proliferation, and migration of human umbilical vein endothelial cells (HUVECs). Furthermore, the effects of BSS treatment on vascular endothelial growth factor receptor 2 (VEGFR2) were determined using molecular docking and Western blotting. Additionally, in the presence or absence of BSS, synovial angiogenesis and joint destruction of the ankle were investigated in collagen-induced arthritis (CIA) mice. The effect of BSS treatment on VEGFR2/p-VEGFR2 expression was verified through immunohistochemical staining.Results: The immunohistochemistry results revealed that BSS treatment inhibited angiogenesis both in vitro and in vivo. In addition, the results of 5-ethynyl-2′-deoxyuridine and cell cycle analysis showed that BSS treatment suppressed the proliferation of HUVECs, while the Transwell migration and stress fiber assays demonstrated that BSS treatment inhibited the migration of HUVECs. Notably, the inhibitory effect of BSS treatment on VEGFR2/p-VEGFR2 was similar to that of axitinib. In CIA mice, BSS also exerted therapeutic effects on the ankles by reducing the degree of swelling, ameliorating bone and cartilage damage, preventing synovial angiogenesis, and inhibiting VEGFR2 and p-VEGFR2 expression.Conclusion: Therefore, our findings demonstrate that BSS exerts an inhibitory effect on synovial angiogenesis by suppressing the proliferation and migration of endothelial cells, thereby alleviating joint swelling and bone destruction in CIA mice. Furthermore, the underlying therapeutic mechanisms may involve the inhibition of VEGF signaling pathway activation.
Collapse
Affiliation(s)
- Kai Qian
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Postdoctoral Research Station, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Xue-Xia Zheng
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Xiao-Bao Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shu-Di Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan-Kai Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min-Ying Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Chang-Song Lin, ; Min-Ying Liu,
| | - Chang-Song Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rheumatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Chang-Song Lin, ; Min-Ying Liu,
| |
Collapse
|