1
|
Liao H, Zheng J, Lu J, Shen HL. NF-κB Signaling Pathway in Rheumatoid Arthritis: Mechanisms and Therapeutic Potential. Mol Neurobiol 2025; 62:6998-7021. [PMID: 39560902 DOI: 10.1007/s12035-024-04634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that imposes a heavy economic burden on patients and society. Bone and cartilage destruction is considered an important factor leading to RA, and inflammation, oxidative stress, and mitochondrial dysfunction are closely related to bone erosion and cartilage destruction in RA. Currently, there are limitations in the clinical treatment methods for RA, which urgently necessitates finding new effective treatments for patients. Nuclear transcription factor-κB (NF-κB) is a signaling transcription factor that is widely present in various cells. It plays an important role as a stress source in the cellular environment and regulates gene expression in processes such as immunity, inflammation, cell proliferation, and apoptosis. NF-κB has long been recognized as a pathogenic factor of RA, and its activation can exacerbate RA by promoting inflammation, oxidative stress, mitochondrial dysfunction, and bone destruction. Conversely, inhibiting the activity of the NF-κB pathway effectively inhibits these pathological processes, thereby alleviating RA. Therefore, NF-κB may be a potential therapeutic target for RA. This article describes the physiological structure of NF-κB and its important role in RA through the regulation of oxidative stress, inflammatory response, mitochondrial function, and bone destruction. Meanwhile, we also summarized the impact of NF-κB crosstalk with other signaling pathways on RA and the effect of related drugs or inhibitors targeting NF-κB on RA. The purpose of this article is to provide evidence for the role of NF-κB in RA and to emphasize its significant role in RA by elucidating the mechanisms, so as to provide a theoretical basis for targeting the NF-κB pathway as a treatment for RA.
Collapse
Affiliation(s)
- Haiyang Liao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jianxiong Zheng
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jinyue Lu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Hai-Li Shen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
2
|
Han X, Gao Y, He M, Luo Y, Wei Y, Duan Y, Zhang S, Yu H, Kan J, Hou T, Zhang Y, Li Y. Evolocumab prevents atrial fibrillation in rheumatoid arthritis rats through restraint of PCSK9 induced atrial remodeling. J Adv Res 2024; 61:211-221. [PMID: 37709197 PMCID: PMC11258665 DOI: 10.1016/j.jare.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
INTRODUCTION Proprotein convertase subtilisin/kexin type 9 (PCSK9) is implicated in the pathogenesis and progression of autoimmune disease. Patients with rheumatoid arthritis (RA) are at high risk of developing atrial fibrillation (AF), while whether PCSK9 is involved in the onset of AF among RA patients remains unclear. OBJECTIVES To explore the role of PCSK9 in the occurrence of AF in RA patients and decipher the underlying mechanism. METHODS We established a rat model of collagen-induced arthritis (CIA) by immunization with type II collagen in Freund's incomplete adjuvant. Atrial electrophysiological test was used to evaluate AF susceptibility. We performed a clinical study to examine the correlation between PCSK9 level and AF, which recruited healthy control, RA patients and RA patients complicated with AF. Evolocumab (a monoclonal antibody of PCSK9) is administered via intraperitoneal injection in CIA rats to assess the role of PCSK9 in RA-related AF. LPS-RS (LPS inhibitor), clodronate liposomes (depletion of macrophages), and cell co-culture model were used to dissect the mechanism underlying PCSK9 promotes AF. RESULTS AF inducibility and duration were higher in CIA rats, accompanied by elevated plasma and atrial PCSK9. Interestingly, compared with healthy control subjects, patients with RA showed an increase in PCSK9, and the PCSK9 is much higher in RA patients complicated with AF. The level of PCSK9 was independently associated with AF risk in RA patients. In the in vivo experiment, evolocumab reduced AF susceptibility, and ameliorated atrial structural remodeling of CIA rats. Mechanistically, augmented LPS in CIA rats led to an increase in PCSK9, which exacerbated fibrosis of cardiac fibroblasts and apoptosis of cardiac myocytes by enhancement of M1 macrophages polarization and inflammation, thereby contributing to AF. CONCLUSION This study suggests that elevated PCSK9 causes atrial structural remodeling by enhancement of M1 macrophages polarization in atria, and evolocumab can effectively protects CIA rats from AF.
Collapse
Affiliation(s)
- Xuejie Han
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yunlong Gao
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Meijiao He
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yingchun Luo
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Ying Wei
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yu Duan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Song Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Hui Yu
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Jiuxu Kan
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Te Hou
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Yun Zhang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China.
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin 150001, China; NHC Key Laboratory of Cell Translation, Harbin Medical University, Heilongjiang 150001, China; Key Laboratory of Hepatosplenic Surgery, Harbin Medical University, Ministry of Education, Harbin 150001, China; Key Laboratory of Cardiac Diseases and Heart Failure, Harbin Medical University, Harbin 150001, China; Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, Harbin 150081, China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, China.
| |
Collapse
|
3
|
Taguchi K, Fukami K. RAGE signaling regulates the progression of diabetic complications. Front Pharmacol 2023; 14:1128872. [PMID: 37007029 PMCID: PMC10060566 DOI: 10.3389/fphar.2023.1128872] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Diabetes, the ninth leading cause of death globally, is expected to affect 642 million people by 2040. With the advancement of an aging society, the number of patients with diabetes having multiple underlying diseases, such as hypertension, obesity, and chronic inflammation, is increasing. Thus, the concept of diabetic kidney disease (DKD) has been accepted worldwide, and comprehensive treatment of patients with diabetes is required. Receptor for advanced glycation endproducts (RAGE), a multiligand receptor, belonging to the immunoglobulin superfamily is extensively expressed throughout the body. Various types of ligands, including advanced glycation endproducts (AGEs), high mobility group box 1, S100/calgranulins, and nucleic acids, bind to RAGE, and then induces signal transduction to amplify the inflammatory response and promote migration, invasion, and proliferation of cells. Furthermore, the expression level of RAGE is upregulated in patients with diabetes, hypertension, obesity, and chronic inflammation, suggesting that activation of RAGE is a common denominator in the context of DKD. Considering that ligand–and RAGE–targeting compounds have been developed, RAGE and its ligands can be potent therapeutic targets for inhibiting the progression of DKD and its complications. Here, we aimed to review recent literature on various signaling pathways mediated by RAGE in the pathogenesis of diabetic complications. Our findings highlight the possibility of using RAGE–or ligand–targeted therapy for treating DKD and its complications.
Collapse
|
4
|
Shabbir MA, Mehak F, Khan MR, Ahmed W, Nawaz MF, Hassoun A, Bhat ZF, Aadil RM. Unraveling the role of natural functional oils in modulating osteoarthritis related complications. Crit Rev Food Sci Nutr 2023; 64:6881-6901. [PMID: 36762672 DOI: 10.1080/10408398.2023.2176815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Osteoarthritis (OA) is a common joint disease and has been studied extensively in recent years as no promising therapy available so far for its treatment and remains a great challenge for health care specialists. Although the identification of some major mechanisms that contribute to this disease suggests a plethora of bioactive agents in tackling the associated complications yet OA's pathophysiology is still poorly understood owing to complex mechanistic changes observed. Experimental research is now exploring a wide range of therapeutically effective agents in an effort to find a way to repair OA-related joint degeneration and halt it from getting worse. Data was acquired and reviewed from most relevant and recent studies. This review summarizes the studies that are currently available and focuses on how various unconventional functional oils affect osteoarthritis and the affected joint tissues. An analysis of the recent scientific literature allowed us to highlight the potential anti-arthritic properties of edible oils and their main constituents, which seems to suggest an interesting new potential therapeutic application. Due to eccentric nature of OA, it is necessary to concentrate initially on the management of symptoms. The evidence supporting functional oils chondroprotective potential is still accumulating, underpinning a global need for more sustainable natural sources of treatment. More clinical research that focuses on the consequences of long-term treatment, possible negative effects, and epigenetic implications is necessary to get optimistic results. However, different animal or clinical studies suggest that linolenic and linoleic fatty acids decreased chondrocyte oxidative stress, cartilage breakdown, and expression of inflammatory markers. Distinct fatty acids along with minor components of oils also reduced the generation of prostaglandins and decreased oxidative stress. Furthermore, the potential roles of the main components of edible oils and possible negative results (if any) are also reported. While no severe side effects have been reported for any edible oils. Overall, these studies identify and support the use of functional oils as an adjuvant therapy for the management of OA and as a means of symptomatic alleviation for OA patients. However, to prove the effectiveness or to draw precise conclusions, high-quality clinical trials are required.
Collapse
Affiliation(s)
- Muhammad Asim Shabbir
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Fakiha Mehak
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Moazzam Rafiq Khan
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Waqar Ahmed
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Furqan Nawaz
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Abdo Hassoun
- Univ. Littoral Côte d'Opale, UMRt 1158 BioEcoAgro, USC ANSES, INRAe, Univ. Artois, Univ. Lille, Univ. Picardie Jules Verne, Univ. Liège, Junia, France
- Sustainable AgriFoodtech Innovation & Research (SAFIR), Arras, France
| | - Zuhaib F Bhat
- Division of Livestock Products Technology, SKUAST-J, Jammu, J&K, India
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| |
Collapse
|
5
|
Sun F, Hao W, Meng X, Xu D, Li X, Zheng K, Yu Y, Wang D, Pan W. Polyene phosphatidylcholine ameliorates synovial inflammation: involvement of PTEN elevation and glycolysis suppression. Mol Biol Rep 2023; 50:687-696. [PMID: 36370296 DOI: 10.1007/s11033-022-08043-3] [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: 05/06/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Synovial inflammation, characterized by the activation of synovial fibroblasts (SFs), is a crucial factor to drive the progression of rheumatoid arthritis (RA). Polyene phosphatidylcholine (PPC), the classic hepatoprotective drug, has been reported to ameliorate arthritis in animals. However, the molecular mechanism remains poorly understood. METHODS AND RESULTS: Using in vitro primary synovial fibroblast (SFs) culture system, we revealed that phosphatase and tension homolog deleted on chromosome 10 (PTEN), a tumor suppressor, mediates the anti-inflammatory effect of PPC in lipopolysaccharide (LPS)-stimulated primary SFs. PPC decreased the production of TNF-α and IL-6 production while elevating the level of IL-10 and TGF-β. Furthermore, PPC up-regulated the expression of PTEN, but inhibited the expression of p-AKT (ser473) and PI3K-p85α. Moreover, pre-treatment of SF1670 (the inhibitor of PTEN) or 740Y-P (the agonist of AKT/PI3K pathways) partially abrogated the anti-inflammatory effect of PPC. In addition, PPC could inhibit the expression of GLUT4, a key transporter of glucose that fuels the glycolysis, which is accompanied by the expression downregualtion of glycolytic enzymes PFKFB3 and PKM2. Furthermore, PPC could reduce ROS production and mitochondrial membrane potential in LPS-stimulated SFs and MH7A cell line. CONCLUSION The present study supported that PPC can alleviate synovial inflammation, which involves in the elevation of PTEN and blockage of glycolysis.
Collapse
Affiliation(s)
- Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,National Experimental Teaching Demonstration Center of Basic Medicine (Xuzhou Medical University), Xuzhou, Jiangsu, China
| | - Wenting Hao
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xianran Meng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangyang Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, Zhejiang, China.
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
6
|
Hypoxia-induced HMGB1 promotes glioma stem cells self-renewal and tumorigenicity via RAGE. iScience 2022; 25:104872. [PMID: 36034219 PMCID: PMC9399482 DOI: 10.1016/j.isci.2022.104872] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/10/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Glioma stem cells (GSCs) in the hypoxic niches contribute to tumor initiation, progression, and recurrence in glioblastoma (GBM). Hypoxia induces release of high-mobility group box 1 (HMGB1) from tumor cells, promoting the development of tumor. Here, we report that HMGB1 is overexpressed in human GBM specimens. Hypoxia promotes the expression and secretion of HMGB1 in GSCs. Furthermore, silencing HMGB1 results in the loss of stem cell markers and a reduction in self-renewal ability of GSCs. Additionally, HMGB1 knockdown inhibits the activation of RAGE-dependent ERK1/2 signaling pathway and arrests the cell cycle in GSCs. Consistently, FPS-ZM1, an inhibitor of RAGE, downregulates HMGB1 expression and the phosphorylation of ERK1/2, leading to a reduction in the proliferation of GSCs. In xenograft mice of GBM, HMGB1 knockdown inhibits tumor growth and promotes mouse survival. Collectively, these findings uncover a vital function for HMGB1 in regulating GSC self-renewal potential and tumorigenicity. Glioma stem cells overexpress HMGB1 in human glioblastoma Hypoxia induces the upregulation and release of HMGB1 in glioma stem cells HMGB1 promotes the self-renewal of glioma stem cells via RAGE Targeting HMGB1 inhibits the tumorigenesis of glioma stem cells
Collapse
|
7
|
CgHMGB1 functions as a broad-spectrum recognition molecule to induce the expressions of CgIL17-5 and Cgdefh2 via MAPK or NF-κB signaling pathway in Crassostrea gigas. Int J Biol Macromol 2022; 211:289-300. [PMID: 35525493 DOI: 10.1016/j.ijbiomac.2022.04.166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/15/2022] [Accepted: 04/22/2022] [Indexed: 01/03/2023]
Abstract
High-mobility group box 1 (HMGB1), a highly conserved nucleoprotein, functions in immune recognition, inflammation and antibacterial immunization in vertebrates. In the present study, the mediation mechanism of CgHMGB1 in activating MAPK and NF-κB/Rel signaling pathways to induce the expressions of immune effectors was investigated. CgHMGB1 mRNA was detected in all tested developmental stages from fertilized egg to D-larvae, with the higher expressions in 4-cell and 8-cell stages. CgHMGB1 proteins were mainly distributed in haemocyte granulocytes. The expressions of CgHMGB1 mRNA in haemocytes increased significantly after Vibrio splendidus stimulation, and CgHMGB1 protein translocated into the haemocyte cytoplasm and release into cell-free haemolymph. The phosphorylation of CgERK and CgP38 were induced, the nuclear translocation of CgRel were promoted, and the mRNA expressions of CgIL17-5 and Cgdefh2 increased significantly after rCgHMGB1 treatment. Obvious branchial swelling and cilium shedding were observed after rCgHMGB1 treatment. rCgHMGB1 exhibited binding activity to different polysaccharides, bacteria, and fungi. rCgHMGB1 also displayed obvious antibacterial activity to V. splendidus and E. coli. These results indicated that CgHMGB1 functioned as an immune recognition molecule to recognize various PAMPs and bacteria to induce the mRNA expressions of CgIL17-5 and Cgdefh2 via the activation of MAPK and NF-κB signaling pathways in oysters.
Collapse
|
8
|
Cai L, Huang N, Zhang X, Wu S, Wang L, Ke Q. Long non-coding RNA plasmacytoma variant translocation 1 and growth arrest specific 5 regulate each other in osteoarthritis to regulate the apoptosis of chondrocytes. Bioengineered 2022; 13:13680-13688. [PMID: 35706414 PMCID: PMC9275885 DOI: 10.1080/21655979.2022.2063653] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) and growth arrest specific 5 (GAS5) have opposite functions in the apoptosis of chondrocytes, which are involved in the pathogenesis of osteoarthritis (OA). The opposite roles of PVT1 and GAS5 in OA may indicate the existence of crosstalk between them in OA. This study aimed to explore the possible interaction between PVT1 and GAS5 in OA. Accumulation of PVT1 and GAS5 in OA and control synovial fluid samples was measured by RT-qPCR. The interaction between PVT1 and GAS5 in chondrocytes was explored by overexpression experiments. Dual-luciferase reporter assay was performed to analyze the binding of PVT1 and GAS5 to each other’s promoter regions. Regulatory roles of PVT1 and GAS5 in the apoptosis of chondrocytes were studied with cell apoptosis assay. PVT1 was upregulated in OA, and GAS5 was downregulated in OA. An inverse correlation between PVT1 and GAS5 was observed across OA samples. Under lipopolysaccharides (LPS) treatment, PVT1 was upregulated and GAS5 was downregulated. Interestingly, PVT1 and GAS5 overexpression downregulated each other in chondrocytes. Cell apoptosis analysis showed that PVT1 overexpression promoted cell apoptosis, while GAS5 overexpression suppressed cell apoptosis induced by LPS. Co-transfection of PVT1 and GAS5 failed to significantly affect cell apoptosis. PVT1 and GAS5 directly bound to each other’s promoter regions. Our study characterized the interaction between PVT1 and GAS5 in OA. Their interaction regulated the apoptosis of chondrocytes, which play a critical role in OA. PVT1 and GAS5 may form a negative feedback loop in OA.
Collapse
Affiliation(s)
- Liquan Cai
- Department of Trauma Orthopaedics, Joint Surgery, Donghai Hospital, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Nianlai Huang
- Department of Trauma Orthopaedics, Joint Surgery, Donghai Hospital, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Xiaolu Zhang
- Department of Trauma Orthopaedics, Joint Surgery, Donghai Hospital, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Shiqiang Wu
- Department of Trauma Orthopaedics, Joint Surgery, Donghai Hospital, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Liangming Wang
- Department of Trauma Orthopaedics, Joint Surgery, Donghai Hospital, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| | - Qingfeng Ke
- Department of Trauma Orthopaedics, Joint Surgery, Donghai Hospital, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, China
| |
Collapse
|
9
|
Chen M, Zhou Y, Xue M, Zhu R, Jing L, Lin L, He C, Qin Y. Anti-HMGB1 antibody is a potential characteristic autoantibody for Sjögren's syndrome. Sci Rep 2022; 12:6020. [PMID: 35411013 PMCID: PMC9001690 DOI: 10.1038/s41598-022-10007-3] [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: 10/15/2021] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Sjögren's syndrome (SS) is a common chronic inflammatory autoimmune disease that affects about 0.33–0.77% population in China. The positive for antinuclear antibodies (ANA) is one of the key features of SS, which shows a nuclear fine speckled (AC-4) pattern in an indirect immunofluorescent antibody test (IIFT). About 70% of ANA-positive SS patients have detectable anti-SS-A and/or SS-B antibodies, which indicates that other autoantibodies may present in SS patients. The anti-HMGB1 antibodies in 93 SS patients and 96 healthy controls were investigated with in-house developed ELISA and immunoblotting, and the locations of HMGB1 and fluorescent pattern of anti-HMGB1 antibody were investigated with IIFT. The contribution of anti-HMGB1 antibody in ANA-IF was evaluated with Cas9-induce HMGB1 knockout B16 cells. The anti-HMGB1 antibody level is higher in SS patients (9.96 ± 5.55 RU/ml) than in healthy controls (4.9 ± 1.4 RU/ml). With ROC curve analysis, when taking 8 RU/ml as the cutoff value, the sensitivity, specificity, and the area under the curve were 64.5%, 96.9%, and 0.83, respectively. A total of 18 patients (20.7%) with nuclear fine speckled (AC-4) pattern in ANA-IF test were anti-HMGB1 antibody positive only. With commercial antibody, anti-HMGB1 antibody showed the same nuclear fine speckled (AC-4) pattern. The serum from ANA-IF (+), SS-A (−), and SS-B (−) SS patients showed nuclear fine speckled (AC-4) pattern in wildtype B16 cells, but no fluorescence in HMGB1 knockout B16 cells. Anti-HMGB1 antibody may be one of the characteristic autoantibodies of SS in addition to anti-SS-A and SS-B. The detection of anti-HMGB1 antibody can provide more laboratory evidence for clinical diagnosis of SS.
Collapse
|
10
|
de Sire A, Marotta N, Marinaro C, Curci C, Invernizzi M, Ammendolia A. Role of Physical Exercise and Nutraceuticals in Modulating Molecular Pathways of Osteoarthritis. Int J Mol Sci 2021; 22:5722. [PMID: 34072015 PMCID: PMC8198532 DOI: 10.3390/ijms22115722] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling disease that affects millions of patients. Its etiology is largely unknown, but it is most likely multifactorial. OA pathogenesis involves the catabolism of the cartilage extracellular matrix and is supported by inflammatory and oxidative signaling pathways and marked epigenetic changes. To delay OA progression, a wide range of exercise programs and naturally derived compounds have been suggested. This literature review aims to analyze the main signaling pathways and the evidence about the synergistic effects of these two interventions to counter OA. The converging nutrigenomic and physiogenomic intervention could slow down and reduce the complex pathological features of OA. This review provides a comprehensive picture of a possible signaling approach for targeting OA molecular pathways, initiation, and progression.
Collapse
Affiliation(s)
- Alessandro de Sire
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| | - Nicola Marotta
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| | - Cinzia Marinaro
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| | - Claudio Curci
- Physical Medicine and Rehabilitation Unit, Department of Neurosciences, ASST Carlo Poma, 46100 Mantova, Italy;
| | - Marco Invernizzi
- Physical Medicine and Rehabilitation, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy;
- Translational Medicine, Dipartimento Attività Integrate Ricerca e Innovazione (DAIRI), Azienda Ospedaliera S.S. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Antonio Ammendolia
- Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy; (N.M.); (C.M.); (A.A.)
| |
Collapse
|
11
|
Wang XP, Xie WP, Bi YF, Wang BA, Song HB, Wang SL, Bi RX. Quercetin suppresses apoptosis of chondrocytes induced by IL-1β via inactivation of p38 MAPK signaling pathway. Exp Ther Med 2021; 21:468. [PMID: 33767763 PMCID: PMC7976442 DOI: 10.3892/etm.2021.9899] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The objective of the present study was to investigate the effect of quercetin and evaluate its protective effect on articular cartilage in patients with osteoarthritis (OA), by intervening the p38 pathway. The target factors of quercetin protecting articular cartilage in patients with OA were predicted scientifically and analyzed to predict the possible pathways by using network pharmacology. A pathway predicted to be closely associated with osteoarthritis was chosen for experimental verification in in vitro cells. The optimal intervention drug concentrations were selected by the of Cell Cycle Kit-8 assay, osteoarthritis and inflammatory factors relevant to osteoarthritis, interleukin-1β and tumor necrosis factor-α, were tested by of enzyme-linked immunosorbent assay, and the expression of relevant proteins and mRNA of the p38 signaling pathway was tested by reverse transcription-quantitative PCR and western blotting, following quercetin intervention. It was found that quercetin, at the concentration of 100 umol/l, can decrease inflammatory factors relevant to OA, inhibit the expression of p38, matrix metalloprotease 13 and ADAMTS in the pathway, and promote the expression of collagen Ⅱ. Therefore, it is postulated that quercetin can lower the expression of inflammatory factors in cartilage for the prevention and treatment of OA, and the expression level of relevant factors can be changed positively by blocking the p38 MAPK signaling pathway. Thus, quercetin can promote the repair of degenerative chondrocytes and protect articular chondrocytes.
Collapse
Affiliation(s)
- Xiang-Peng Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Wen-Peng Xie
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Yi-Fei Bi
- Special Inspection Department, Affiliated Ophthalmic Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250002, P.R. China
| | - Bao-An Wang
- Shandong Institute of Commerce and Technology, Jinan, Shandong 250103, P.R. China
| | - Hong-Bo Song
- College of Foreign Languages, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355 P.R. China
| | - Shi-Lu Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China.,Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| | - Rong-Xiu Bi
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China.,Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
12
|
Chen M, Zhu L, Xue M, Zhu R, Jing L, Wang H, Qin Y. HMGB1, anti-HMGB1 antibodies, and ratio of HMGB1/anti-HMGB1 antibodies as diagnosis indicator in fever of unknown origin. Sci Rep 2021; 11:5059. [PMID: 33658546 PMCID: PMC7930274 DOI: 10.1038/s41598-021-84477-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/16/2021] [Indexed: 11/29/2022] Open
Abstract
To evaluate the feasibility of serum HMGB1, anti-HMGB1 antibodies, and HMGB1/anti-HMGB1 ratio as a diagnosis indicator of initial clinical classification in patients with fever of unknown origin (FUO). Ninety-four patients with classical FUO and ninety healthy controls were enrolled in this study. The subjects’ clinical data and serum were collected. The serum concentration of HMGB1 was detected by a commercial HMGB1 ELISA kit, while the serum concentration of anti-HMGB1 antibodies were detected by an in-house built anti-HMGB1 antibodies ELISA kit and further confirmed by immunoblotting. According to the hospital diagnosis on discharge, ninety-four FUO patients were divided into four groups, Infectious disease subgroup, autoimmune disease subgroup, malignant tumor subgroup, and undetermined subgroup. The concentrations of HMGB1 in the infectious disease subgroup and autoimmune disease subgroup were higher than those in the malignant tumor subgroup, undetermined subgroup, and healthy control group. The concentration of anti-HMGB1 antibodies in autoimmune disease subtype group was higher than those in other subgroups as well as healthy control group. According to the distribution of HMGB1 and anti-HMGB1 in scatter plots of the patients with FUO, we found that the ratio of serum HMGB1/anti-HMGB1 is an ideal clinical indicator for differential diagnosis of different subtypes of FUO. The best cut-off was 0.75, and the sensitivity, specificity, and AUC were 66.67%, 87.32%, and 0.8, respectively. Correlation analysis showed that serum concentration of HMGB1 was moderately correlated with CRP in infectious diseases subgroup, and the serum concentration of anti-HMGB1 antibodies was strongly correlated with erythrocyte sedimentation rate in autoimmune disease subgroup. Our study had showed that serum HMGB1/anti-HMGB1 antibodies ratio can help clinicians identify FUO subtypes, thereby avoiding many unnecessary examinations and tests, and improving the effectiveness of clinical diagnosis and treatment of FUO.
Collapse
Affiliation(s)
- Mingkun Chen
- Department of Laboratory Medicine, Changhai Hospital, SMMU, Shanghai, China
| | - Li Zhu
- Department of Laboratory Medicine, Wuxi People's Hospital, Wuxi, China
| | - Miao Xue
- Department of Laboratory Medicine, Changhai Hospital, SMMU, Shanghai, China
| | - Rongrong Zhu
- Department of Laboratory Medicine, Changhai Hospital, SMMU, Shanghai, China
| | - Liling Jing
- Department of Laboratory Medicine, Changhai Hospital, SMMU, Shanghai, China
| | - Huaizhou Wang
- Department of Laboratory Medicine, Changhai Hospital, SMMU, Shanghai, China.
| | - Yanghua Qin
- Department of Laboratory Medicine, Changhai Hospital, SMMU, Shanghai, China.
| |
Collapse
|
13
|
He YJ, Liang X, Zhang XX, Li SS, Sun Y, Li TF. PTH1-34 inhibited TNF-α expression and antagonized TNF-α-induced MMP13 expression in MIO mice. Int Immunopharmacol 2021; 91:107191. [PMID: 33359852 DOI: 10.1016/j.intimp.2020.107191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 10/22/2022]
Abstract
This study aims to investigate the effects and mechanisms of parathyroid hormone [1-34] (PTH1-34) on TNF-α-stimulated mice chondrocytes, as well as cartilage from a meniscus injury induced osteoarthritis (MIO) mice model. The C57BL/6J mice received medial meniscectomy, and then administrated with PTH1-34. The results showed that PTH1-34 administration decreased secondary allodynia and the pain-related transcripts. The IHC, ELISA, Micro-CT imaging and histopathology analysis revealed the significantly improved subchondral plate thickness and bone porosity, the reduced pro-inflammatory cytokines in serum and joint fluid. In vitro, mice chondrocyte was treated with TNF-α or co-cultured with synovial cells. The results showed that TNF-α markedly upregulated the MMP13 expression, and the ERK1/2, NF-κB or PI3K signaling pathway inhibitors could reverse the induction effect of TNF-α on expression of MMP13 in chondrocytes. PTH1-34 alone has no effect on the expression of MMP13 and NF-κB signaling pathways, but the PTH1-34 could reverse the induction effect of TNF-α on MMP13 expression and NF-κB signaling pathway activation in chondrocytes. In addition, PTH1-34 administration inhibited the expression of TNF-α and MMP13, and chondrocyte viability, while the PKA repressor reversed the effect of PTH1-34 in chondrocytes co-cultured with synovial cells. In conclusion, PTH1-34 has an obvious analgesic and anti-inflammatory effect, inhibits the matrix synthesis and alleviates the progression of osteoarthritis. In vitro, PTH1-34 inhibited TNF-α expression and antagonized TNF-α-induced MMP13 expression via the PKA pathway and the NF-κB signaling pathways, respectively.
Collapse
Affiliation(s)
- Yu-Jie He
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xu Liang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xin-Xin Zhang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Shan-Shan Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yue Sun
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Tian-Fang Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
14
|
Wang Y, Gao W. Effects of TNF-α on autophagy of rheumatoid arthritis fibroblast-like synoviocytes and regulation of the NF-κB signaling pathway. Immunobiology 2021; 226:152059. [PMID: 33561598 DOI: 10.1016/j.imbio.2021.152059] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/20/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is a common chronic autoimmune disease, which seriously harms human health. The hyperplastic growth of fibroblast-like synoviocytes (FLSs) plays a key role in the pathogenesis of RA. However, the pathogenesis of RA remains unclear. In this experiment, we confirmed that Tumor necrosis factor alpha (TNF-α) could activate the autophagy of RA-FLSs. 3-Methyladenine (3-MA) and Chloroquine (CQ), two types of autophagy blocker, combined with TNF-α were used to treat FLSs. The results showed that this treatment caused a reduction in the level of autophagy-related protein, significant increases in the expression of apoptosis-related protein and the apoptosis rate, and significant inhibition of the proliferation-promoting ability of TNF-α. Ammonium pyrrolidinedithiocarbamate (PDTC), a specific nuclear factor kappa-B (NF-κB) activity blocker, significantly inhibited autophagy induced by TNF-α. Collectively, these findings showed, for the first time, that TNF-α can up-regulate autophagy activity and activate the NF-κB signal pathway. Inhibition of autophagy can improve the imbalance of proliferation/apoptosis of FLSs aggravated by TNF-α to some extent, thus delaying the progression of RA. The NF-κB signal pathway may be involved in the regulation of FLSs autophagy by TNF-α.
Collapse
Affiliation(s)
- Yu Wang
- Department of Rheumatology, the First Affiliated Hospital of Jinzhou Medical University, Liaoning, Jinzhou 121000, China
| | - Wei Gao
- Department of Rheumatology, the First Affiliated Hospital of Jinzhou Medical University, Liaoning, Jinzhou 121000, China.
| |
Collapse
|
15
|
Saiyang X, Qingqing W, man X, Chen L, Min Z, Yun X, Wenke S, Haiming W, Xiaofeng Z, Si C, Haipeng G, Wei D, Qizhu T. Activation of Toll-like receptor 7 provides cardioprotection in septic cardiomyopathy-induced systolic dysfunction. Clin Transl Med 2021; 11:e266. [PMID: 33463061 PMCID: PMC7775988 DOI: 10.1002/ctm2.266] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND As a pattern recognition receptor, Toll-like receptor 7 (TLR7) widely presented in the endosomal membrane of various cells. However, the precise role and mechanism of TLR7 in septic cardiomyopathy remain unknown. This study aims to determine the role of TLR7 in cardiac dysfunction during sepsis and explore the mechanism of TLR7 in septic cardiomyopathy. METHODS We generated a mouse model of septic cardiomyopathy by challenging with lipopolysaccharide (LPS). TLR7-knockout (TLR7-/- ), wild-type (WT) mice, cardiac-specific TLR7-transgenic (cTG-TLR7) overexpression, and littermates WT (LWT) mice were subjected to septic model. Additionally, to verify the role and mechanism of TLR7 in vitro, we transfected neonatal rat ventricular myocytes (NRVMs) with Ad-TLR7 and TLR7 siRNA before LPS administration. The effects of TLR7 were assessed by Ca2+ imaging, western blotting, immunostaining, and quantitative real-time polymerase chain reaction (qPCR). RESULTS We found that TLR7 knockout markedly exacerbated sepsis-induced systolic dysfunction. Moreover, cardiomyocytes isolated from TLR7-/- mice displayed weaker Ca2+ handling than that in WT mice in response to LPS. Conversely, TLR7 overexpression alleviated LPS-induced systolic dysfunction, and loxoribine (TLR7-specific agonist) improved LPS-induced cardiac dysfunction. Mechanistically, these optimized effects were associated with enhanced the adenosine (cAMP)-protein kinase A (PKA) pathway, which upregulated phosphorylate-phospholamban (p-PLN) (Ser16) and promoted sarco/endoplasmic reticulum Ca2+ ATPase (Serca) and Ryanodine Receptor 2 (RyR2) expression in the sarcoplasmic reticulum (SR), and ultimately restored Ca2+ handling in response to sepsis. While improved Ca2+ handling was abrogated after H89 (a specific PKA inhibitor) pretreatment in cardiomyocytes isolated from cTG-TLR7 mice. Consistently, TLR7 overexpression improved LPS-induced Ca2+ -handling decrement in NRVMs. Nevertheless, TLR7 knockdown showed a deteriorative phenotype. CONCLUSIONS Our data demonstrated that activation of TLR7 protected against sepsis-induced cardiac dysfunction through promoting cAMP-PKA-PLN pathway, and we revealed that TLR7 might be a novel therapeutic target to block the septic cardiomyopathy and support systolic function during sepsis.
Collapse
Affiliation(s)
- Xie Saiyang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Wu Qingqing
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Xu man
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Liu Chen
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Zhang Min
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Xing Yun
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Shi Wenke
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Wu Haiming
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Zeng Xiaofeng
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Chen Si
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| | - Guo Haipeng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of HealthQilu Hospital of Shandong UniversityJinanChina
- Department of Critical Care MedicineQilu Hospital of Shandong UniversityJinanPeople's Republic of China
| | - Deng Wei
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
- Department of CardiologyThe Fifth Affiliated Hospital of Xinjiang Medical UniversityÜrümqiChina
| | - Tang Qizhu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanPeople's Republic of China
| |
Collapse
|
16
|
Xue J, Suarez JS, Minaai M, Li S, Gaudino G, Pass HI, Carbone M, Yang H. HMGB1 as a therapeutic target in disease. J Cell Physiol 2020; 236:3406-3419. [PMID: 33107103 DOI: 10.1002/jcp.30125] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/13/2020] [Indexed: 12/30/2022]
Abstract
High-mobility group box 1 (HMGB1) was initially recognized as a ubiquitous nuclear protein involved in maintaining the nucleosome integrity and facilitating gene transcription. HMGB1 has since been reevaluated to be a prototypical damage-associated molecular pattern (DAMP) protein, and together with its exogenous counterpart, pathogen-associated molecular pattern (PAMP), completes the body's alarmin system against disturbances in homeostasis. HMGB1 can be released into the extracellular matrix (ECM) by either granulocytes or necrotic cells to serve as a chemotaxis/cytokine during infection, endotoxemia, hypoxia, ischemia-reperfusion events, and cancer. Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Targeting HMGB1 constitutes a favorable therapeutic strategy for inflammation and inflammatory diseases. Antagonists such as ethyl pyruvate inhibit HMGB1 by interfering with its cytoplasmic exportation, while others such as glycyrrhizin directly bind to HMGB1 and render it unavailable for its receptors. The fact that a mixture of different HMGB1 isoforms is present in the ECM poses a challenge in pinpointing the exact role of an individual antagonist. A more discriminative probe for HMGB1 may be necessary to advance our knowledge of HMGB1, HMGB1 antagonists, and inflammatory-related diseases.
Collapse
Affiliation(s)
- Jiaming Xue
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA.,John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Joelle S Suarez
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Michael Minaai
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Shuangjing Li
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA.,Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Giovanni Gaudino
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York, USA
| | - Michele Carbone
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Haining Yang
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| |
Collapse
|
17
|
Meng Y, Qiu S, Sun L, Zuo J. Knockdown of exosome‑mediated lnc‑PVT1 alleviates lipopolysaccharide‑induced osteoarthritis progression by mediating the HMGB1/TLR4/NF‑κB pathway via miR‑93‑5p. Mol Med Rep 2020; 22:5313-5325. [PMID: 33174011 PMCID: PMC7646997 DOI: 10.3892/mmr.2020.11594] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis is a chronic degenerative joint disease. Long non‑coding RNA plasmacytoma variant translocation 1 (PVT1) is involved in the progression of osteoarthritis and exosomes serve a central role in intercellular communication. However, whether PVT1 can be mediated by exosomes in osteoarthritis has not been reported. Whole blood was drawn from osteoarthritis patients and healthy volunteers. Lipopolysaccharide (LPS) was used to stimulate human normal chondrocytes (C28/I2) to construct a cell damage model in vitro. Protein levels were examined via western blot analysis. eThe expression of PVT1, microRNA (miR)‑93‑5p and high mobility groupprotein B1 (HMGB1) was evaluated through reverse transcription‑quantitative PCR. Cell viability and apoptosis were determined through CCK‑8 or flow cytometric assay. Inflammatory cytokines were measured via ELISA. The relationship between PVT1 or HMGB1 and miR‑93‑5p was confirmed by dual‑luciferase reporter assay. PVT1, HMGB1 and exosomal PVT1 were upregulated while miR‑93‑5p was downregulated in osteoarthritis patient serum and LPS‑induced C28/I2 cells. Exosomes from osteoarthritis patient serum and LPS‑treated C28/I2 cells increased PVT1 expression in C28/I2 cells. PVT1 depletion reversed the decrease of viability and the increase of apoptosis, inflammation responses and collagen degradation of C28/I2 cells induced by LPS. PVT1 regulated HMGB1 expression via sponging miR‑93‑5p. miR‑93‑5p inhibition abolished PVT1 silencing‑mediated viability, apoptosis, inflammation responses and collagen degradation of LPS‑stimulated C28/I2 cells. HMGB1 increase overturned miR‑93‑5p upregulation‑mediated viability, apoptosis, inflammation responses and collagen degradation of LPS‑stimulated C28/I2 cells. Furthermore, PVT1 modulated the Toll‑like receptor 4/NF‑κB pathway through an miR‑93‑5p/HMGB1 axis. In summary, exosome‑mediated PVT1 regulated LPS‑induced osteoarthritis progression by modulating the HMGB1/TLR4/NF‑κB pathway via miR‑93‑5p, providing a new route for possible osteoarthritis treatment.
Collapse
Affiliation(s)
- Yong Meng
- Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Siqiang Qiu
- Department of Spine Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| | - Long Sun
- Department of Orthopedics, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Jinliang Zuo
- Department of Spine Surgery, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
18
|
Han D, Fang Y, Tan X, Jiang H, Gong X, Wang X, Hong W, Tu J, Wei W. The emerging role of fibroblast-like synoviocytes-mediated synovitis in osteoarthritis: An update. J Cell Mol Med 2020; 24:9518-9532. [PMID: 32686306 PMCID: PMC7520283 DOI: 10.1111/jcmm.15669] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/29/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA), the most ubiquitous degenerative disease affecting the entire joint, is characterized by cartilage degradation and synovial inflammation. Although the pathogenesis of OA remains poorly understood, synovial inflammation is known to play an important role in OA development. However, studies on OA pathophysiology have focused more on cartilage degeneration and osteophytes, rather than on the inflamed and thickened synovium. Fibroblast-like synoviocytes (FLS) produce a series of pro-inflammatory regulators, such as inflammatory cytokines, nitric oxide (NO) and prostaglandin E2 (PGE2 ). These regulators are positively associated with the clinical symptoms of OA, such as inflammatory pain, joint swelling and disease development. A better understanding of the inflammatory immune response in OA-FLS could provide a novel approach to comprehensive treatment strategies for OA. Here, we have summarized recently published literatures referring to epigenetic modifications, activated signalling pathways and inflammation-associated factors that are involved in OA-FLS-mediated inflammation. In addition, the current related clinical trials and future perspectives were also summarized.
Collapse
Affiliation(s)
- Dafei Han
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Yilong Fang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Xuewen Tan
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Haifei Jiang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Xun Gong
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Xinming Wang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Wenming Hong
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Jiajie Tu
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Gacaferi H, Mimpen JY, Baldwin MJ, Snelling SJB, Nelissen RGHH, Carr AJ, Dakin SG. The potential roles of high mobility group box 1 (HMGB1) in musculoskeletal disease: A systematic review. TRANSLATIONAL SPORTS MEDICINE 2020. [DOI: 10.1002/tsm2.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Hamez Gacaferi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
- Department of Orthopaedics Leiden University Medical Centre Leiden The Netherlands
| | - Jolet Y. Mimpen
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Mathew J. Baldwin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Sarah J. B. Snelling
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | | | - Andrew J. Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Stephanie G. Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| |
Collapse
|
20
|
Cheng M, Wang Y. Downregulation of HMGB1 by miR-103a-3p Promotes Cell Proliferation, Alleviates Apoptosis and in Flammation in a Cell Model of Osteoarthritis. IRANIAN JOURNAL OF BIOTECHNOLOGY 2020; 18:e2255. [PMID: 32884953 PMCID: PMC7461710 DOI: 10.30498/ijb.2020.129470.2255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background MiR-103a-3p is a small non-coding RNA and has been reported to be involved in osteogenic proliferation and differentiation, but the role of miR-103a-3p in human osteoarthritis (OA) remains unclear. Objectives In this study, we aimed to explore its function and molecular target in chondrocytes during OA pathogenesis. Materials and Methods Total 12 experimental OA rat models, together with 12 rats without knee OA lesions were established and cartilage samples were collected. Chondrocytes were treated with LPS in vitro. MiR-103a-3p expression was detected in articular cartilage tissues and chondrocytes using quantitative real-time PCR. Knee OA chondrocytes were transfected with miR-103a-3p mimics, and siHMGB1, respectively. Then cellular proliferation, apoptosis, apoptosis related factors and inflammatory cytokines were analyzed by MTT, flow cytometry, western blot, caspase-3 activity and ELISA, respectively. Potential targets of miR-103a-3p were predicted using series of bioinformatics analysis, then confirmed by luciferase reporter assay. Results We first found miR-103a-3p was significantly down-regulated in the articular cartilage tissues from experimental OA rats, as well as in chondrocytes treated with LPS in vitro. The gain-of-function assay further demonstrated that up-regulation of miR-103a-3p significantly promoted cell proliferation, inhibited apoptosis and inflammation, which was accompanied with elevated expression of PCNA, and reduced expression of caspase-3, PARP, IL-1β, IL-6, IL-10 and TNF-α. Furthermore, high mobility group box 1 (HMGB1), an important inflammatory mediator of OA, was a target of miR-103a-3p. Moreover, knockdown of HMGB1 mimicked the effects of miR-103a-3p on chondrocytes treated with LPS. Conclusions Taken together, our study suggests that miR-103a-3p inhibits chondrocyte apoptosis and inflammation in OA, which appears to be an attractive approach to OA treatment.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Rehabilitation, Jinniu District People's Hospital of Chengdu, Chengdu, Sichuan, China.,Department of Orthopaedics, Sichuan Academy of Medical Sciences & Sichuan Provincial people's Hospital, Chengdu, Sichuan, China
| | - Yue Wang
- Department of Orthopaedics, Sichuan Academy of Medical Sciences & Sichuan Provincial people's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Gorgulho CM, Romagnoli GG, Bharthi R, Lotze MT. Johnny on the Spot-Chronic Inflammation Is Driven by HMGB1. Front Immunol 2019; 10:1561. [PMID: 31379812 PMCID: PMC6660267 DOI: 10.3389/fimmu.2019.01561] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Although much has been made of the role of HMGB1 acting as an acute damage associated molecular pattern (DAMP) molecule, prompting the response to tissue damage or injury, it is also released at sites of chronic inflammation including sites of infection, autoimmunity, and cancer. As such, the biology is distinguished from homeostasis and acute inflammation by the recruitment and persistence of myeloid derived suppressor cells, T regulatory cells, fibrosis and/or exuberant angiogenesis depending on the antecedents and the other individual inflammatory partners that HMGB1 binds and focuses, including IL-1β, CXCL12/SDF1, LPS, DNA, RNA, and sRAGE. High levels of HMGB1 released into the extracellular milieu and its persistence in the microenvironment can contribute to the pathogenesis of many if not all autoimmune disorders and is a key factor that drives inflammation further and worsens symptoms. HMGB1 is also pivotal in the maintenance of chronic inflammation and a “wound healing” type of immune response that ultimately contributes to the onset of carcinogenesis and tumor progression. Exosomes carrying HMGB1 and other instructive molecules are released and shape the response of various cells in the chronic inflammatory environment. Understanding the defining roles of REDOX, DAMPs and PAMPs, and the host response in chronic inflammation requires an alternative means for positing HMGB1's central role in limiting and focusing inflammation, distinguishing chronic from acute inflammation.
Collapse
Affiliation(s)
- Carolina M Gorgulho
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil.,DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Graziela G Romagnoli
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Rosh Bharthi
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael T Lotze
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
22
|
Li H, Liu P, Gong Y, Liu J, Ruan F. Expression and function of miR-155 in rat synovial fibroblast model of rheumatoid arthritis. Exp Ther Med 2019; 18:786-792. [PMID: 31258713 DOI: 10.3892/etm.2019.7581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 03/01/2018] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by joint synovial inflammation and is a challenge for researchers and clinicians. MicroRNAs (miRNAs/miRs) represent a group of small non-coding RNA molecules that post-transcriptionally regulate mRNA expression and are involved in various diseases, including cancer, autoimmune and metabolic diseases, as well as neurological disorders. In the present study, various experiments were performed to investigate the effects and underlying mechanism of miR-155 in RA using rat synoviocytes induced by lipopolysaccharide (LPS) to model rheumatoid arthritis. It was revealed that synovial fibroblasts exhibited significantly higher miR-155 mRNA levels than the control group. Compared with the RA group, the viability of synovial fibroblasts was significantly decreased in the miR-155 mimics + RA group, but markedly increased in the miR-155 inhibitor + RA group. Compared with that in the RA + NC mimic or RA + NC inhibitor groups, the apoptosis of synovial fibroblasts increased significantly in the miR-155 mimics + RA group, but was significantly decreased in the miR-155 inhibitor + RA group. The miR-155 mimics + RA group exhibited higher expression levels of β-catenin, matrix metalloproteinase 7 and cyclin D1 compared with the miR-155 inhibitor + RA group, and the glycogen synthase kinase protein levels was lower compared with the miR-155 inhibitor + RA group. In brief, it was inferred that the Wnt signaling pathway is involved in the miR-155-associated inhibition of RA synovial fibroblast viability and induction of cell apoptosis. Inhibition of miR-155 may be an effective treatment for RA through regulation of the Wnt signaling pathway to reduce cell apoptosis and enhance cell viability.
Collapse
Affiliation(s)
- Hewei Li
- Department of Orthopedics, Liyuan Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Ping Liu
- Department of Orthopedics, Liyuan Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Yanlin Gong
- Department of Endocrinology, Wuhan No. 1 Hospital, Wuhan, Hubei 430022, P.R. China
| | - Jiali Liu
- Department of Orthopedics, Liyuan Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| | - Feng Ruan
- Department of Orthopedics, Liyuan Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430077, P.R. China
| |
Collapse
|
23
|
Novel Hybrid Gels Made of High and Low Molecular Weight Hyaluronic Acid Induce Proliferation and Reduce Inflammation in an Osteoarthritis In Vitro Model Based on Human Synoviocytes and Chondrocytes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4328219. [PMID: 31179322 PMCID: PMC6507116 DOI: 10.1155/2019/4328219] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/07/2019] [Accepted: 04/01/2019] [Indexed: 12/22/2022]
Abstract
High molecular weight hyaluronan (H-HA) has a pivotal role in the maintenance of normal functions of synovial fluid and structure of the articular joint, but it has been shown that its concentration is reduced in patients affected by degenerative cartilage diseases, such as osteoarthritis (OA). The aim of this study was to investigate the anti-inflammatory effects and properties of hybrid cooperative complexes based on high and low molecular weight hyaluronan (HCC) compared to H-HA on human primary cells derived by pathological joints. In addition, the rheological behavior of HCC was evaluated in order to define their potential as viscosupplement gel in degenerated joints. The experiments were performed using an in vitro model of OA based on human chondrocytes and synoviocytes isolated from degenerated joints of patients hospitalized for surgical replacement. In order to assess the anti-inflammatory effects of HCC, we evaluated NF-kB, COMP-2, IL-6, and IL-8 as specific markers at the transcriptional and/or protein level. Moreover, the proliferative properties of HCC were assessed using time lapse video microscopy. We showed that chondrocytes and synoviocytes clearly presented an altered cytokine profile compatible with a severe ongoing inflammation status. H-HA and, above all, HCC significantly reduced levels of the specific biomarkers evaluated and improved cartilage healing. The rheological profile indicated HCC suitability for intra-articular injection in joint diseases. HCC viscoelastic properties and the protective/anti-inflammatory effect on human chondrocytes and synoviocytes suggest the novel HCC-based gels as a valid support for OA management.
Collapse
|
24
|
Zhang T, Guan XW, Gribben JG, Liu FT, Jia L. Blockade of HMGB1 signaling pathway by ethyl pyruvate inhibits tumor growth in diffuse large B-cell lymphoma. Cell Death Dis 2019; 10:330. [PMID: 30988279 PMCID: PMC6465275 DOI: 10.1038/s41419-019-1563-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023]
Abstract
High mobility group box 1 (HMGB1) protein in the tumor microenvironment actively contributes to tumor progression but its role in diffuse large B-cell lymphoma (DLBCL) is unknown. The aim of this study was to determine the mechanism by which HMGB1 promotes tumor growth in DLBCL and whether blockade of HMGB1 signaling pathway could inhibit tumorigenesis. We report that HMGB1 promotes proliferation of DLBCL cells by activation of AKT, extracellular signal-regulated kinases 1/2 (ERK1/2), signal transducer and activator of transcription 3 (STAT3) and SRC Proto-Oncogene, Non-Receptor Tyrosine Kinase (Src). Ethyl pyruvate (EP), an anti-inflammatory agent, inhibits HMGB1 active release from DLBCL cells and significantly inhibited proliferation of DLBCL cells in vitro. Treatment with EP significantly prevented and inhibited tumor growth in vivo and prolonged DLBCL-bearing mice survival. EP significantly downregulated HMGB1 expression and phosphorylation of Src and ERK1/2 in mice lymphoma tissue. EP induced accumulation of the cell cycle inhibitor p27 but downregulated expression of cyclin-dependent kinase 2 (CDK2). Increased nuclear translocation of p27 interacted with CDK2 and cyclin A, which led to blockade of cell cycle progression at the G1 to S phase transition. In conclusion, we demonstrated for the first time that blockade of HMGB1-mediated signaling pathway by EP effectively inhibited DLBCL tumorigenesis and disease progression.
Collapse
Affiliation(s)
- Tian Zhang
- Department of Radiotherapy, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xu-Wen Guan
- Department of Radiotherapy, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin Medical University, Tianjin, China
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Feng-Ting Liu
- Department of Radiotherapy, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China. .,Department of Hematology and Oncology, Tianjin Union Medical Center, Tianjin, China.
| | - Li Jia
- Department of Radiotherapy, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China. .,Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| |
Collapse
|
25
|
Lema C, Reins RY, Redfern RL. High-Mobility Group Box 1 in Dry Eye Inflammation. Invest Ophthalmol Vis Sci 2019; 59:1741-1750. [PMID: 29610858 PMCID: PMC5886030 DOI: 10.1167/iovs.17-23363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose To determine high-mobility group box 1 (HMGB1) expression during experimental dry eye (EDE) and dry eye-like culture conditions and elucidate its role in corneal dry eye-related inflammation. Methods EDE was induced in 8- to 12-week-old C57BL/6 mice. Corneal tissue sections and lysates from EDE and untreated mice were evaluated for HMGB1 expression by immunostaining and quantitative real-time PCR (qPCR). For in vitro studies, human corneal epithelial cells (HCEC) were treated with hyperosmolar media, toll-like receptor (TLR) agonists, or proinflammatory cytokines to determine HMGB1 expression. HCEC were also treated with human recombinant HMGB1 (hrHMGB1) alone or in combination with inflammatory stimuli, and TNFα, IL-6, and IL-8 expression evaluated by qPCR and ELISA. Nuclear factor-κB (NF-κB) p65 nuclear translocation was determined by immunostaining. Results EDE mice had higher corneal HMGB1 RNA and protein expression compared to untreated animals. In HCEC, hyperosmolar stress and TNFα treatment stimulated HMGB1 production and secretion into culture supernatants. However, in vitro stimulation with hrHMGB1 did not induce secretion of TNFα, IL-6, or IL-8 or NF-κB p65 nuclear translocation. In addition, the inflammatory response elicited by TLR agonists fibroblast-stimulating lipopeptide-1 and lipopolysaccharide was not enhanced by hrHMGB1 treatment. Conclusions HMGB1 expression was enhanced by dry eye conditions in vivo as well as in vitro, during hyperosmolar stress and cytokine exposure, suggesting an important role for HMGB1 in dry eye disease. However, no direct inflammatory effect was observed with HMGB1 treatment. Therefore, under these conditions, HMGB1 does not contribute directly to dry eye-induced inflammation and its function at the ocular surface needs to be explored further.
Collapse
Affiliation(s)
- Carolina Lema
- The Ocular Surface Institute, University of Houston, College of Optometry, Houston, Texas, United States
| | - Rose Y Reins
- The Ocular Surface Institute, University of Houston, College of Optometry, Houston, Texas, United States
| | - Rachel L Redfern
- The Ocular Surface Institute, University of Houston, College of Optometry, Houston, Texas, United States
| |
Collapse
|
26
|
Zhang W, Wu J, Zhang F, Dou X, Ma A, Zhang X, Shao H, Zhao S, Ling P, Liu F, Han G. Lower range of molecular weight of xanthan gum inhibits apoptosis of chondrocytes through MAPK signaling pathways. Int J Biol Macromol 2019; 130:79-87. [PMID: 30659877 DOI: 10.1016/j.ijbiomac.2019.01.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/14/2019] [Accepted: 01/16/2019] [Indexed: 01/04/2023]
Abstract
LRWXG has previously been reported to have a protective effect on chondrocytes, preventing apoptosis induced by oxidative stress. In this study, we were aimed at determining whether LRWXG exerts its anti-apoptotic activity through the MAPK signaling pathways in chondrocytes. Our results show that, at the cellular level, apoptosis of chondrocytes in the groups treated by LRWXG decreases compared with groups treated by inhibitors alone and model group under conditions of oxidative stress in a dose-dependent manner. Mechanistically at the molecular level, LRWXG regulates the MAPK pathway induced by oxidative stress: The levels of phosphorylation of JNK and p38 proteins in the groups treated by LRWXG are lower than model group, while compared with corresponding groups of inhibitors, there are no significant difference; For other related proteins, LRWXG reduces the levels of the apoptosis-related proteins BAX and cleaved caspase-3, and increases the level of anti-apoptotic protein BCL2. In addition, LRWXG can significantly reduce the levels of inflammatory-related factors such as COX2, PEG2, TNFα and IL1β, and inhibits the expression of MMPs, increasing the content of type II collagen. The results of this research strongly suggest that LRWXG exerts its anti-apoptotic activity via regulating the MAPK signaling pathways in vitro.
Collapse
Affiliation(s)
- Wei Zhang
- Jinzhou Medical University, Jinzhou 121001, China; The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China; Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Jixu Wu
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Fangfang Zhang
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Xixi Dou
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Aibin Ma
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Xiaogang Zhang
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Huarong Shao
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Shuo Zhao
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Peixue Ling
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Fei Liu
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China.
| | - Guanying Han
- Jinzhou Medical University, Jinzhou 121001, China; The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China.
| |
Collapse
|
27
|
Zhang C, Yu W, Huang C, Ding Q, Liang C, Wang L, Hou Z, Zhang Z. Chrysin protects human osteoarthritis chondrocytes by inhibiting inflammatory mediator expression via HMGB1 suppression. Mol Med Rep 2018; 19:1222-1229. [PMID: 30535473 DOI: 10.3892/mmr.2018.9724] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 09/19/2018] [Indexed: 11/05/2022] Open
Abstract
High‑mobility group box chromosomal protein (HMGB‑1) contributes to osteoarthritis (OA) by modulating various oxidative, inflammatory and apoptotic signaling pathways. The effect of chrysin (CH), a natural plant flavonoid, and its functional interaction with HMGB‑1, was investigated in a chondrocyte model of OA. Human chondrocytes were pre‑treated with CH, and then subsequently treated with IL‑1β to induce the formation of chondrocytes similar to those found in OA joints. Next, the expression level of HMGB‑1 was determined by immunofluorescence and western blot analysis. Additionally, inflammatory factor expression was measured by ELISA, and cell apoptosis was analyzed with flow cytometry. To further explore the effects of CH, HMGB‑1 expression was silenced following CH treatment with small interfering (si)RNA. The results demonstrated that CH inhibited cell apoptosis, dose‑dependently reduced matrix metalloproteinase (MMP) 13, collagenase and IL‑6 expression, and increased collagen α‑1 (II) chain (COL2A1) expression in human osteoarthritis chondrocytes. These effects of CH were accompanied by decreased HMGB‑1 expression. Additionally, the expression of MMP13, collagenase, IL‑6 and COL2A1, as well as apoptosis, was significantly reduced by HMGB‑1 siRNA. These results demonstrated that HMGB‑1 is critical for the protective effect of CH on human osteoarthritis chondrocytes, including cell apoptosis and inflammatory factor inhibition, which suggests that CH may have potential therapeutic effect in treating OA by protecting human osteoarthritis chondrocytes via HMGB1 suppression.
Collapse
Affiliation(s)
- Chi Zhang
- Orthopedic Department, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Weizhong Yu
- Department of Knee Surgery and Sport Medicine, Guangzhou Orthopedic Hospital, Guangzhou, Guangdong 510045, P.R. China
| | - Chongbo Huang
- Department of Knee Surgery and Sport Medicine, Guangzhou Orthopedic Hospital, Guangzhou, Guangdong 510045, P.R. China
| | - Qinghe Ding
- Department of Knee Surgery and Sport Medicine, Guangzhou Orthopedic Hospital, Guangzhou, Guangdong 510045, P.R. China
| | - Chizhang Liang
- Department of Knee Surgery and Sport Medicine, Guangzhou Orthopedic Hospital, Guangzhou, Guangdong 510045, P.R. China
| | - Le Wang
- Orthopedic Department, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Zhiqi Hou
- Department of Knee Surgery and Sport Medicine, Guangzhou Orthopedic Hospital, Guangzhou, Guangdong 510045, P.R. China
| | - Zhiyong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing Technologies of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| |
Collapse
|
28
|
van den Bosch MHJ. Inflammation in osteoarthritis: is it time to dampen the alarm(in) in this debilitating disease? Clin Exp Immunol 2018; 195:153-166. [PMID: 30421798 DOI: 10.1111/cei.13237] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2018] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease that strongly reduces the quality of life in patients; However, no disease-modifying therapy is available. For a long time, OA was considered a non-inflammatory disease that was the result of 'wear-and-tear' and abnormal mechanics, and therefore many considered the term 'osteoarthritis' a misnomer. However, during the last decades the notion arose that inflammation is not only present in the majority of OA patients but, rather, actively involved in the progression of the disease. Influx of immune cells is observed in the synovium and a plethora of inflammatory mediators is present in tissues and fluids from OA patients. These mediators cause the production of degrading enzymes that break down the cartilage matrix, which is the main hallmark of OA. Alarmins, which belong to the group of danger signals, have been implicated in many inflammatory diseases. They are among the first factors to be released upon cell stress due to, for example, infection, damage and inflammation. They attract and activate cells of the immune system and therefore lie at the base of the inflammatory reaction. In this narrative review, an overview of the history of OA, the evolving concept of inflammation as important factor in the OA pathogenesis, and particularly the central role that alarmins play in the initiation and maintenance of the low-grade inflammatory response in OA, is provided. Moreover, the targeting of alarmins as a promising approach to dampen the inflammation in OA is highlighted.
Collapse
Affiliation(s)
- M H J van den Bosch
- Experimental Rheumatology, Radboud university medical center, Nijmegen, the Netherlands
| |
Collapse
|
29
|
Ashour H, Rashed LA, El-Sebaie MM, Sabry MM, Abdelmohsen MA, Eissa M. Combined gemfibrozil (peroxisome proliferator-activated receptor alpha agonist) with reduced steroid dose gives a similar management picture as the full steroid dose in a rat adjuvant-induced arthritis model. Mod Rheumatol 2018; 29:602-611. [PMID: 30074417 DOI: 10.1080/14397595.2018.1508800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Objectives: The study aimed to evaluate the efficacy of combined gemfibrozil with prednisolone in the management of adjuvant-induced arthritis (AIA) rat model. Methods: Seventy two adult male Wistar albino rats were divided equally into 1-control group, three diseased groups: 2- Adjuvant induced arthritis (AIA), 3- high fat diet (HF), and 4- combined AIA-HF, and treated groups: 5- gemfibrozil 30 mg/kg treated AIA group (AIA-G) and the combined AIA-HF treated groups: 6- prednisolone equivalent to human 10 mg (AIA-HF-P10), 7- prednisolone equivalent to human 5 mg (AIA-HF-P5) 8- gemfibrozil (HF-AIA-G) and 9- combined treatment (HF-AIA-G-P5) Results: HF diet represents a precipitating factor for knee arthritis. Gemfibrozil improved the inflammatory findings in both AIA and AIA-HF groups. Combined administration of gemfibrozil with reduced steroid dose gave a similar therapeutic effect to the full steroid dose. Fortunately, we reported more reduction in the nuclear factor kappa-B (NF-κB) and high mobility group box 1 (HMGB1) in the HF-AIA-G-P5 compared with the HF-AIA-P10 group. The improvement was proved by the histological findings. Conclusion: Combined reduced prednisolone dose with gemfibrozil potentiates its anti-inflammatory activity. This could be a target in the management of rheumatoid arthritis.
Collapse
Affiliation(s)
- Hend Ashour
- a Department of Physiology, Kasr Alainy, Faculty of Medicine , Cairo University , Cairo , Egypt
| | - Laila Ahmed Rashed
- b Department of Biochemistry, Kasr Alainy, Faculty of Medicine , Cairo University , Cairo , Egypt
| | | | - Marwa Mohammed Sabry
- c Department of Histology, Kasr Alainy, Faculty of Medicine , Cairo University , Cairo , Egypt
| | - Manal Ali Abdelmohsen
- c Department of Histology, Kasr Alainy, Faculty of Medicine , Cairo University , Cairo , Egypt
| | - Mervat Eissa
- d Department of Rheumatology, Kasr Alainy, Faculty of Medicine , Cairo University , Cairo , Egypt
| |
Collapse
|
30
|
Zhang L, Luan L, Ma Y. Dishevelled‑2 modulates osteogenic differentiation of human synovial fibroblasts in osteoarthritis. Mol Med Rep 2018; 18:292-298. [PMID: 29749449 PMCID: PMC6059681 DOI: 10.3892/mmr.2018.8975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 03/29/2018] [Indexed: 12/25/2022] Open
Abstract
Dishevelled (Dvl)-2 represents one of the cytoplasmic proteins, which serves as a pivotal hub in signaling intermediates through a number of different signaling pathways associated with the Wnt family. The aim of the present study was to investigate the roles and mechanisms of Dvl-2 on synovial fibroblasts (SFBs) in osteoarthritis (OA). A Cell Counting kit-8 (CCK-8) assay was used to determine cell viability. An alkaline phosphatase (ALP) test kit was used to measure the activity of ALP. Western blot and reverse transcription-quantitative polymerase chain reaction analysis were used to evaluate the protein and mRNA expression, respectively. The results suggest that depletion of Dvl-2 significantly decreased the expression of osteoprotegerin (OPG) and ALP (P<0.05) and significantly increased the expression of receptor activator of nuclear factor-κB ligand (RANKL), ALP, osteonectin (ON), osteocalcin (OCN) and osterix (P<0.05). In addition, the depletion of Dvl-2 also significantly inhibited the expression of runt-related transcription factor 2 (Runx-2) and β-catenin in SFBs (P<0.05). The effect of Dvl-2 over-expression was opposite to the effect of Dvl-2 silencing. The inactivation of Wnt3a reversed the effect of Dvl-2 silencing. In conclusion, the results indicate that Dvl-2 regulated osteogenic differentiation of SFBs in OA.
Collapse
Affiliation(s)
- Lihua Zhang
- Department of Rheumatology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Luan Luan
- Department of Rheumatology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| | - Yingying Ma
- Department of Rheumatology, Jining No. 1 People's Hospital, Jining, Shandong 272000, P.R. China
| |
Collapse
|
31
|
Chimenti MS, Perricone C, Novelli L, Caso F, Costa L, Bogdanos D, Conigliaro P, Triggianese P, Ciccacci C, Borgiani P, Perricone R. Interaction between microbiome and host genetics in psoriatic arthritis. Autoimmun Rev 2018; 17:276-283. [PMID: 29378263 DOI: 10.1016/j.autrev.2018.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022]
|
32
|
Cai J, Yuan SX, Yang F, Tao QF, Yang Y, Xu QG, Wang ZG, Yu J, Lin KY, Wang ZY, Ma JZ, Zhou CC, Wang F, Sun SH, Zhou WP. Paraoxonase 3 inhibits cell proliferation and serves as a prognostic predictor in hepatocellular carcinoma. Oncotarget 2018; 7:70045-70057. [PMID: 27661119 PMCID: PMC5342533 DOI: 10.18632/oncotarget.12145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022] Open
Abstract
Paraoxonase 3 (PON3) exerts prominent anti-inflammation and anti-oxidation properties mainly at the cellular level, and is primarily expressed in the liver. However, its role in HCC remains unexplored. Here, we investigated the expression pattern, clinical significance, and function of PON3 in HCC. PON3 mRNA and protein levels were respectively determined in two large cohorts using quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) of tissue microarray. We found that PON3 was downregulated in most HCCs. Kaplan-Meier and log-rank test showed that PON3 downregulation predicted shorter recurrence-free survival (RFS) and overall survival (OS) time in all HCC patients, especially early-stage HCC patients. Cox regression analysis revealed that the PON3 downregulation was an independent risk factor for RFS and OS. Gain- and loss-of-function experiments revealed that PON3 suppressed cell proliferation in vivo and in vitro, which was attributed to its cell-cycle arrest effect. In addition, microarray analysis showed that some pro-proliferative genes were elevated when PON3 was knockdown, and these genes possibly involved in the underlying mechanisms. In conclusion, our studies reveal the cell proliferation inhibitory function of PON3 and offer a potential prognostic predictor and therapeutic target for HCC.
Collapse
Affiliation(s)
- Jie Cai
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Sheng-Xian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Fu Yang
- The Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | - Qi-Fei Tao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Qing-Guo Xu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Zhen-Guang Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jian Yu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Kong-Ying Lin
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Zong-Yan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jin-Zhao Ma
- The Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | - Chuan-Chuan Zhou
- The Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | - Fang Wang
- The Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | - Shu-Han Sun
- The Department of Medical Genetics, Second Military Medical University, Shanghai, China
| | - Wei-Ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
33
|
BAY 11-7085 induces glucocorticoid receptor activation and autophagy that collaborate with apoptosis to induce human synovial fibroblast cell death. Oncotarget 2018; 7:23370-82. [PMID: 26993765 PMCID: PMC5029633 DOI: 10.18632/oncotarget.8042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 02/28/2016] [Indexed: 11/29/2022] Open
Abstract
Inhibition of proapoptotic pathways in synovial fibroblasts is one of the major causes of synovial proliferation and hyperplasia in rheumatic diseases. We have shown previously that NF-κB inhibitor BAY 11-7085, through inactivation of PPAR-γ, induces apoptosis in human synovial fibroblasts. In this work we showed that BAY 11-7085 induced autophagy that preceded BAY 11-7085-induced apoptosis. Of interest, BAY 11-7085 induced Serine 211 phosphorylation and degradation of glucocorticoid receptor (GR). Glucocorticoid prednisolone induced both activation and degradation of GR, as well as autophagy in synovial fibroblasts. BAY 11-7085-induced cell death was significantly decreased with glucocorticoid inhibitor mifepristone and with inhibitors of autophagy. Both BAY 11-7085-induced autophagy and GR activation were down regulated with PPAR-γ agonist, 15d-PGJ2 and MEK/ERK inhibitor UO126. Inhibition of autophagy markedly decreased endogenous and BAY 11-7085-induced ERK phosphorylation, suggesting a positive feed back loop between ERK activation and autophagy in synovial fibroblasts. Co-transfection of MEK1 with PPAR-γ1 in HEK293 cells caused known inhibitory phosphorylation of PPAR-γ1 (Serine 112) and enhanced GR degradation, in the absence or presence of prednisolone. Furthermore, GR was both phosphorylated on Serine 211 and down regulated in synovial fibroblasts during serum starvation induced autophagy. These results showed that GR activation and PPAR-γ inactivation mediated BAY 11-7085-induced autophagy.
Collapse
|
34
|
BRD4 has dual effects on the HMGB1 and NF-κB signalling pathways and is a potential therapeutic target for osteoarthritis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3001-3015. [DOI: 10.1016/j.bbadis.2017.08.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/28/2017] [Accepted: 08/16/2017] [Indexed: 01/11/2023]
|
35
|
Shimizu K, Kamada Y, Sakamoto A, Matsuda M, Nakatsuka M, Hiramatsu Y. High Expression of High-Mobility Group Box 1 in Menstrual Blood: Implications for Endometriosis. Reprod Sci 2017; 24:1532-1537. [PMID: 29017437 DOI: 10.1177/1933719117692042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Endometriosis is a benign gynecologic disease characterized by the presence of ectopic endometrium and associated with inflammation and immune abnormalities. However, the molecular basis for endometriosis is not well understood. To address this issue, the present study examined the expression of high-mobility group box (HMGB) 1 in menstrual blood to investigate its role in the ectopic growth of human endometriotic stromal cells (ESCs). A total of 139 patients were enrolled in this study; 84 had endometriosis and 55 were nonendometriotic gynecological patients (control). The HMGB1 levels in various fluids were measured by enzyme-linked immunosorbent assay. Expression of receptor for advanced glycation end products (RAGE) in eutopic and ectopic endometrium was assessed by immunohistochemistry, and RAGE and vascular endothelial growth factor ( VEGF) messenger RNA expression in HMGB1- and lipopolysaccharide (LPS)-treated ESCs was evaluated by real-time polymerase chain reaction. The HMGB1 concentration was higher in menstrual blood than in serum or peritoneal fluid ( P < .001 for both). RAGE was expressed in both normal and ectopic endometrium. Administration of 1000 ng/mL HMGB1 or coadministration of 100 ng/mL HMGB1 and 100 ng/mL LPS induced VEGF production in ESCs relative to the control ( P < .05). These results suggest that menstrual fluid has naturally high levels of HMGB1 and may promote endometriosis following retrograde menstruation when complexed with other factors such as LPS by inducing inflammation and angiogenesis.
Collapse
Affiliation(s)
- Keiko Shimizu
- 1 Department of Obstetrics and Gynecology, Okayama University Hospital, Okayama, Japan.,2 Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yasuhiko Kamada
- 1 Department of Obstetrics and Gynecology, Okayama University Hospital, Okayama, Japan
| | - Ai Sakamoto
- 1 Department of Obstetrics and Gynecology, Okayama University Hospital, Okayama, Japan.,2 Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Miwa Matsuda
- 1 Department of Obstetrics and Gynecology, Okayama University Hospital, Okayama, Japan.,2 Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mikiya Nakatsuka
- 1 Department of Obstetrics and Gynecology, Okayama University Hospital, Okayama, Japan.,3 Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Yuji Hiramatsu
- 1 Department of Obstetrics and Gynecology, Okayama University Hospital, Okayama, Japan.,2 Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
36
|
The Autophagy Level Is Increased in the Synovial Tissues of Patients with Active Rheumatoid Arthritis and Is Correlated with Disease Severity. Mediators Inflamm 2017; 2017:7623145. [PMID: 28255205 PMCID: PMC5309404 DOI: 10.1155/2017/7623145] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/11/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022] Open
Abstract
Rheumatoid arthritis (RA) is a complex and not fully understood autoimmune disease associated with multijoint damage. The main effector cells, the synovial fibroblasts, are apoptosis resistant and hyperplastic which indicate that autophagy level is high in synovial tissue. Real-time PCR, immunocytochemistry, and western blotting were used in this paper to study the autophagy status of the synovial tissues obtained from RA and OA patients at the time of joint replacement surgery. We further evaluated the correlation between autophagy levels with RA activity-associated serum markers with SPSS. The results showed that the expression levels (both in mRNA and in protein level) of autophagy-related proteins (belcin1, Atg5, and LC3) in the synovial tissue of patients with active rheumatoid arthritis (n = 20) were significantly higher than those in OA patients (n = 16). We further showed that the LC3-II/β-actin relative gray value was strongly correlated with the serum levels of several RA activity-related markers: CRP, ESR, CCP, and RF. Our results indicate that evaluating the autophagy level of synovial biopsies might be a useful way to diagnose RA and to estimate the disease activity. Reducing the expression level of autophagy-related genes might become a new therapeutic target for active rheumatoid arthritis.
Collapse
|
37
|
Zhang W, Guo S, Li B, Liu L, Ge R, Cao T, Wang H, Gao T, Wang G, Li C. Proinflammatory effect of high-mobility group protein B1 on keratinocytes: an autocrine mechanism underlying psoriasis development. J Pathol 2016; 241:392-404. [PMID: 27859256 DOI: 10.1002/path.4848] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/13/2016] [Accepted: 11/05/2016] [Indexed: 12/16/2022]
Abstract
Psoriasis is an autoimmune skin disease, in which keratinocytes play a crucial pathogenic role. High-mobility group protein B1 (HMGB1) is an inflammatory factor that can be released from keratinocyte nuclei in psoriatic lesions. We aimed to investigate the proinflammatory effect of HMGB1 on keratinocytes and the contribution of HMGB1 to psoriasis development. Normal human keratinocytes were treated with recombinant human HMGB1, and the production of inflammatory factors and the intermediary signalling pathways were examined. Furthermore, the imiquimod-induced psoriasis-like mouse model was used to investigate the role of HMGB1 in psoriasis development in vivo. A total of 11 inflammatory factors were shown to be upregulated by HMGB1 in keratinocytes, among which interleukin (IL)-18 showed the greatest change. We then found that activation of the nuclear factor-κB signalling pathway and inflammasomes accounted for HMGB1-induced IL-18 expression and secretion. Moreover, HMGB1 and downstream IL-18 contributed to the development of psoriasiform dermatitis in the imiquimod-treated mice. In addition, T-helper 17 immune response in the psoriasis-like mouse model could be inhibited by both HMGB1 and IL-18 blockade. Our findings indicate that HMGB1 secreted from keratinocytes can facilitate the production and secretion of inflammatory factors such as IL-18 in keratinocytes in an autocrine way, thus promoting the development of psoriasis. Blocking the proinflammatory function of the HMGB1-IL-18 axis may be useful for psoriasis treatment in the future. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Weigang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Bing Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Lin Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Rui Ge
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Tianyu Cao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi, PR China
| |
Collapse
|
38
|
Ugrinova I, Pasheva E. HMGB1 Protein: A Therapeutic Target Inside and Outside the Cell. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:37-76. [PMID: 28215228 DOI: 10.1016/bs.apcsb.2016.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
High-mobility group box 1 protein (HMGB1) is a nonhistone chromosomal protein discovered more than 30 years ago. It is an abundant nuclear protein that has a dual function-in the nucleus, it binds DNA and participates in practically all DNA-dependent processes serving as an architectural factor. Outside the cell, HMGB1 plays a different role-it acts as an alarmine that activates a large number of HMGB1-"competent" cells and mediates a broad range of physiological and pathological responses. This universality makes it an attractive target for innovative therapeutic strategies in the treatment of various diseases. Here we present an overview of the major nuclear and extracellular properties of HMGB1 and describe its interaction with different molecular partners as specific receptors or inhibitors, which are important for its role as a target in multiple diseases. We highlight its pivotal role as a target for cancer treatment at two aspects: first in terms of its substantial impact on the repair capacity of cancer cells, thus affecting the effectiveness of chemotherapy with the antitumor drug cis-platinum and, second, the possibility to be targeted by microRNAs influencing different pathways of human diseases, thus making it a promising candidate for a new strategy for therapeutic interventions against various pathological conditions but mainly cancer.
Collapse
Affiliation(s)
- I Ugrinova
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | - E Pasheva
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
39
|
Choi HW, Klessig DF. DAMPs, MAMPs, and NAMPs in plant innate immunity. BMC PLANT BIOLOGY 2016. [PMID: 27782807 DOI: 10.1186/s12870-016-0921-232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
BACKGROUND Multicellular organisms have evolved systems/mechanisms to detect various forms of danger, including attack by microbial pathogens and a variety of pests, as well as tissue and cellular damage. Detection via cell-surface receptors activates an ancient and evolutionarily conserved innate immune system. RESULT Potentially harmful microorganisms are recognized by the presence of molecules or parts of molecules that have structures or chemical patterns unique to microbes and thus are perceived as non-self/foreign. They are referred to as Microbe-Associated Molecular Patterns (MAMPs). Recently, a class of small molecules that is made only by nematodes, and that functions as pheromones in these organisms, was shown to be recognized by a wide range of plants. In the presence of these molecules, termed Nematode-Associated Molecular Patterns (NAMPs), plants activate innate immune responses and display enhanced resistance to a broad spectrum of microbial and nematode pathogens. In addition to pathogen attack, the relocation of various endogenous molecules or parts of molecules, generally to the extracellular milieu, as a result of tissue or cellular damage is perceived as a danger signal, and it leads to the induction of innate immune responses. These relocated endogenous inducers are called Damage-Associated Molecular Patterns (DAMPs). CONCLUSIONS This mini-review is focused on plant DAMPs, including the recently discovered Arabidopsis HMGB3, which is the counterpart of the prototypic animal DAMP HMGB1. The plant DAMPs will be presented in the context of plant MAMPs and NAMPs, as well as animal DAMPs.
Collapse
Affiliation(s)
- Hyong Woo Choi
- Boyce Thompson Institute, Cornell University, 533 Tower Road, Ithaca, NY, 14853, USA
| | - Daniel F Klessig
- Boyce Thompson Institute, Cornell University, 533 Tower Road, Ithaca, NY, 14853, USA.
| |
Collapse
|
40
|
Choi HW, Klessig DF. DAMPs, MAMPs, and NAMPs in plant innate immunity. BMC PLANT BIOLOGY 2016; 16:232. [PMID: 27782807 PMCID: PMC5080799 DOI: 10.1186/s12870-016-0921-2] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/19/2016] [Indexed: 05/13/2023]
Abstract
BACKGROUND Multicellular organisms have evolved systems/mechanisms to detect various forms of danger, including attack by microbial pathogens and a variety of pests, as well as tissue and cellular damage. Detection via cell-surface receptors activates an ancient and evolutionarily conserved innate immune system. RESULT Potentially harmful microorganisms are recognized by the presence of molecules or parts of molecules that have structures or chemical patterns unique to microbes and thus are perceived as non-self/foreign. They are referred to as Microbe-Associated Molecular Patterns (MAMPs). Recently, a class of small molecules that is made only by nematodes, and that functions as pheromones in these organisms, was shown to be recognized by a wide range of plants. In the presence of these molecules, termed Nematode-Associated Molecular Patterns (NAMPs), plants activate innate immune responses and display enhanced resistance to a broad spectrum of microbial and nematode pathogens. In addition to pathogen attack, the relocation of various endogenous molecules or parts of molecules, generally to the extracellular milieu, as a result of tissue or cellular damage is perceived as a danger signal, and it leads to the induction of innate immune responses. These relocated endogenous inducers are called Damage-Associated Molecular Patterns (DAMPs). CONCLUSIONS This mini-review is focused on plant DAMPs, including the recently discovered Arabidopsis HMGB3, which is the counterpart of the prototypic animal DAMP HMGB1. The plant DAMPs will be presented in the context of plant MAMPs and NAMPs, as well as animal DAMPs.
Collapse
Affiliation(s)
- Hyong Woo Choi
- Boyce Thompson Institute, Cornell University, 533 Tower Road, Ithaca, NY 14853 USA
| | - Daniel F. Klessig
- Boyce Thompson Institute, Cornell University, 533 Tower Road, Ithaca, NY 14853 USA
| |
Collapse
|
41
|
|
42
|
Chen Y, Zhang J, Wang X, Wu Y, Zhu L, Lu L, Shen Q, Qin Y. HMGB1 level in cerebrospinal fluid as a complimentary biomarker for the diagnosis of tuberculous meningitis. SPRINGERPLUS 2016; 5:1775. [PMID: 27795917 PMCID: PMC5061653 DOI: 10.1186/s40064-016-3478-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 10/05/2016] [Indexed: 01/08/2023]
Abstract
Purpose High mobility group box-1 (HMGB1) is a proinflammatory, DAMP protein that participates in many pathological conditions. In this study, we evaluated the usability of CSF HMGB1 as a biomarker for the diagnosis of tuberculous meningitis (TBM). Methods A total of 59 TBM patients and 169 control patients were included in our study. CSF samples were obtained and analyzed for HMGB1 using a commercial ELISA kit. Results The mean CSF HMGB1 was 19.36 ng/ml in TBM patients (n = 59) versus 3.12 ng/ml in non-TB meningitis patients (n = 30), 2.13 ng/ml in patients with extra neural tuberculosis (n = 73), and 1.06 ng/m in controls (n = 66). According to the receiver operator characteristic curves, a cut-off value of 3.4 ng/ml was calculated, indicating that the sensitivity and specificity of CSF HMGB1 alone in diagnosis of TBM were 61.02 and 89.94 %, respectively. In patients with extra neural tuberculosis and a high risk of TBM, CSF HMGB1 seemed to be a good candidate for early differential diagnosis of TBM at the cut-off value of 3.8 ng/ml, when the sensitivity and specificity were 79.49 and 94.52 % respectively. Conclusion Our finding may prove to be clinically useful, because CSF HMGB1 ELISA can be performed in almost all clinical laboratories, especially when sophisticated technologies are either time consuming or unavailable.
Collapse
Affiliation(s)
- Yan Chen
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072 People's Republic of China.,Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, 200433 People's Republic of China
| | - Jun Zhang
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433 People's Republic of China
| | - Xiaofei Wang
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433 People's Republic of China
| | - Yu Wu
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, 200433 People's Republic of China
| | - Li Zhu
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, 200433 People's Republic of China
| | - Longkun Lu
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, 200433 People's Republic of China
| | - Qian Shen
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, 200433 People's Republic of China
| | - Yanghua Qin
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai, 200433 People's Republic of China
| |
Collapse
|
43
|
Serum HMGB1 Serves as a Novel Laboratory Indicator Reflecting Disease Activity and Treatment Response in Ankylosing Spondylitis Patients. J Immunol Res 2016; 2016:6537248. [PMID: 27800496 PMCID: PMC5069358 DOI: 10.1155/2016/6537248] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/10/2016] [Indexed: 01/10/2023] Open
Abstract
Objective. High mobility group box 1 (HMGB1) is a late inflammatory factor participating in the pathogenesis of various autoimmune and inflammatory diseases. In the current study, we analyzed the association between serum levels of HMGB1 and clinical features of AS patients before and during treatment. Methods. Serum HMGB1 was detected in 147 AS patients and 61 healthy controls using ELISA. We evaluated the association between HMGB1 and extra-articular manifestations as well as disease severity indices. Among these AS patients, 41 patients received close follow-up at 1, 3, and 6 months after treatment. This group comprised 25 patients treated with anti-TNF-α biologics and 16 patients receiving oral NSAIDs plus sulfasalazine. Results. The serum HMGB1 of AS patients was significantly higher than in healthy controls and positively correlated with BASDAI, BASFI, ASDAS-ESR, ASDAS-CRP, ESR, and CRP, but not with HLA-B27, anterior uveitis, and recurrent diarrhea. There was no significant difference between patients with radiographic damage of hip joints and those without. We observed that serum HMGB1 paralleled disease activity after treatment. Conclusion. Serum level of HMGB1 is higher in AS patients, and to some extent, HMGB1 can reflect the activity of AS and be used as a laboratory indicator to reflect the therapeutic response.
Collapse
|
44
|
Wang X, Guo Y, Wang C, Yu H, Yu X, Yu H. MicroRNA-142-3p Inhibits Chondrocyte Apoptosis and Inflammation in Osteoarthritis by Targeting HMGB1. Inflammation 2016; 39:1718-28. [DOI: 10.1007/s10753-016-0406-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
45
|
Huang D, Zhao Q, Liu H, Guo Y, Xu H. PPAR-α Agonist WY-14643 Inhibits LPS-Induced Inflammation in Synovial Fibroblasts via NF-kB Pathway. J Mol Neurosci 2016; 59:544-53. [PMID: 27339772 DOI: 10.1007/s12031-016-0775-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/26/2016] [Indexed: 11/28/2022]
Abstract
Osteoarthritis (OA), the most prevalent form of arthritis that results from breakdown of joint cartilage and underlying bone, has been viewed as a chronic condition manifested by persistence of inflammatory responses and infiltration of lymphocytes. Regulation of the inflammatory responses in synovial fibroblasts might be useful to prevent the development and deterioration of osteoarthritis. WY-14643, a potent peroxisome proliferator activator receptor-α (PPAR-α) agonist, has been described to beneficially regulate inflammation in many mammalian cells. Here, we investigate the potential anti-inflammatory role of WY-14643 in lipopolysaccharide (LPS)-induced synovial fibroblasts. WY-14643 greatly inhibited the production of NO and PGE2 induced by LPS. In addition, the mRNA expression of intracellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), endothelin-1 (ET-1), and tissue factor (TF) was significantly suppressed by WY-14643, as well as the secretion of pro-inflammatory cytokines including interleukin-6 (IL-6), IL-1β, tumor necrosis factor-α (TNF-α), and monocyte chemotactic protein-1 (MCP-1). Furthermore, the transcription activity and nuclear translocation of NF-kB were found to be markedly decreased by WY-14643, while the phosphorylation of IkB was enhanced, indicating that the anti-inflammatory role of WY-14643 was meditated by NF-kB-dependent pathway. The application of WY-14643 failed to carry out its anti-inflammatory function in PPAR-α silenced cells, suggesting the role of PPAR-α. These findings may facilitate further studies investigating the translation of pharmacological PPAR-α activation into clinical therapy of OA.
Collapse
Affiliation(s)
- Degang Huang
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, People's Republic of China.
| | - Quanlai Zhao
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Hongfei Liu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Yongjie Guo
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, People's Republic of China
| | - Hongguang Xu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui, People's Republic of China.
| |
Collapse
|
46
|
Yu B, Li X, Wan Q, Han W, Deng C, Guo C. High-Mobility Group Box-1 Protein Disrupts Alveolar Elastogenesis of Hyperoxia-Injured Newborn Lungs. J Interferon Cytokine Res 2016; 36:159-68. [PMID: 26982166 DOI: 10.1089/jir.2015.0080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although high-mobility group box-1 (HMGB1) levels in tracheal aspirates are associated with the pathological features of bronchopulmonary dysplasia (BPD), the role of HMGB1 in the terminal stage of abnormal alveologenesis has not yet been understood. In this study, we addressed the role of HMGB1 in the elastogenesis disruption in the lungs of newborn mice with BPD. We found that elevations of whole lung HMGB1 level were associated with impaired alveolar development and aberrant elastin production in 85% O2-exposed lungs. HMGB1 neutralizing antibody attenuated the structural disintegration developed in hyperoxia-damaged lungs. Furthermore, HMGB1 inhibition rescued the neutrophil influx in hyperoxia-injured lung and partially abolished the mRNA level of the proinflammatory mediators, interleukin (IL)-1β and transforming growth factor (TGF)-β1. These data suggested that pulmonary HMGB1 plays an important role in the disruption of elastogenesis in the terminal stage of lung development through reduced pulmonary inflammatory response.
Collapse
Affiliation(s)
- Benli Yu
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Xiaoyu Li
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Qiufeng Wan
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Wenli Han
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,3 Department of Pharmacology, Chongqing Medical University , Chongqing, P.R. China
| | - Chun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Chunbao Guo
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,4 Department of Hepatology and Liver Transplantation Center, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| |
Collapse
|
47
|
Ma YH, Ma TT, Wang C, Wang H, Chang DY, Chen M, Zhao MH. High-mobility group box 1 potentiates antineutrophil cytoplasmic antibody-inducing neutrophil extracellular traps formation. Arthritis Res Ther 2016; 18:2. [PMID: 26739852 PMCID: PMC4718033 DOI: 10.1186/s13075-015-0903-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/18/2015] [Indexed: 11/29/2022] Open
Abstract
Background Recent studies found that the circulating high-mobility group box 1 (HMGB1) levels could reflect the disease activity of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). HMGB1 could prime neutrophils by increasing ANCA antigens translocation for ANCA-mediated respiratory burst and degranulation. The current study aimed to investigate whether HMGB1 participates in ANCA-induced neutrophil extracellular traps (NETs) formation, which is one of the most important pathogenic aspects in the development of AAV. Methods NETs were induced by treating neutrophils with HMGB1 and ANCA-positive IgG in vitro. NETs formation was assessed using immunofluorescence microscopy and fluorescence probe. Antagonist for relevant receptors Toll-like receptor (TLR)2, TLR4 and the receptor for advanced glycation end products (RAGE), as well as NADPH oxidase molecules were employed. Results The percentage of NETs formation was significantly higher in neutrophils stimulated with HMGB1 plus ANCA-positive IgG than that in neutrophils incubated with HMGB1 or ANCA-positive IgG alone. Consistently, compared with the nonstimulated neutrophils, the cell-free DNA (cfDNA) concentration of NETs was significantly increased from 334.09 ± 46.89 ng/ml to 563.32 ± 122.07 ng/ml in the neutrophils incubated with HMGB1 plus MPO-ANCA-positive IgG (P < 0.001), and from 303.44 ± 37.14 ng/ml to 563.79 ± 145.94 ng/ml in the neutrophils incubated with HMGB1 plus PR3-ANCA-positive IgG (P < 0.001). The aforementioned effect was significantly attenuated by antagonist for relevant receptors TLR2, TLR4 and RAGE, as well as blocking NADPH oxidase. Conclusions HMGB1 can potentiate ANCA-inducing NETs formation and may be involved in the pathogenesis of AAV. HMGB1 exerts effects on NETs formation through interaction with TLR2, TLR4 and RAGE, and the process is NADPH oxidase dependent. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0903-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yun-Hua Ma
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China. .,Renal Division, Department of Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| | - Tian-Tian Ma
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| | - Chen Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| | - Huan Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| | - Dong-Yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| |
Collapse
|
48
|
Xu K, Cai YS, Lu SM, Li XL, Liu L, Li Z, Liu H, Xu P. Autophagy induction contributes to the resistance to methotrexate treatment in rheumatoid arthritis fibroblast-like synovial cells through high mobility group box chromosomal protein 1. Arthritis Res Ther 2015; 17:374. [PMID: 26702616 PMCID: PMC4718027 DOI: 10.1186/s13075-015-0892-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/08/2015] [Indexed: 11/15/2022] Open
Abstract
Background Rheumatoid arthritis fibroblast-like synovial cells (RA-FLS) show resistance to methotrexate (MTX) treatment. To better understand the mechanisms of this resistance, RA-FLS and osteoarthritis fibroblast-like synovial cells (OA-FLS) were isolated and exposed to MTX. We analyzed the autophagy induced by MTX in vitro and its relationship to apoptosis. Methods Cell viability was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and apoptosis was detected by flow cytometry and Western blot analysis. Autophagy was determined by transmission electron microscopy as well as Western blot analysis. The expression levels of Beclin-1, LC3, Akt, p-Akt, mammalian target of rapamycin (mTOR), p-mTOR, high mobility group box chromosomal protein 1 (HMGB1), and an 85 kDa caspase cleaved fragment of poly(ADP-ribose) polymerase were measured by Western blotting. Results MTX-induced apoptosis was increased in OA-FLS compared with RA-FLS. However, MTX stimulated the autophagy response in RA-FLS by inducing autophagosome formation, but not in OA-FLS. In RA-FLS, transfection with Beclin-1 small interfering RNA inhibited autophagy and increased susceptibility to MTX, which induces cell death. MTX upregulated autophagy through its ability to enhance the expression of HMGB1 and Beclin-1 rather than through the Akt/mTOR pathway. Conclusions Autophagy induction contributes to resistance to MTX treatment in fibroblasts from patients with rheumatoid arthritis. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0892-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710054, Shaanxi Province, China.
| | - Yong-song Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710054, Shaanxi Province, China.
| | - She-Min Lu
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, China.
| | - Xiao-Li Li
- Department of Internal Medicine, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, China.
| | - Lin Liu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710054, Shaanxi Province, China.
| | - Zhong Li
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710054, Shaanxi Province, China.
| | - Hui Liu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710054, Shaanxi Province, China.
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710054, Shaanxi Province, China.
| |
Collapse
|
49
|
The interplay between inflammation and metabolism in rheumatoid arthritis. Cell Death Dis 2015; 6:e1887. [PMID: 26379192 PMCID: PMC4650442 DOI: 10.1038/cddis.2015.246] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 07/29/2015] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by extensive synovitis resulting in erosions of articular cartilage and marginal bone that lead to joint destruction. The autoimmune process in RA depends on the activation of immune cells, which use intracellular kinases to respond to external stimuli such as cytokines, immune complexes, and antigens. An intricate cytokine network participates in inflammation and in perpetuation of disease by positive feedback loops promoting systemic disorder. The widespread systemic effects mediated by pro-inflammatory cytokines in RA impact on metabolism and in particular in lymphocyte metabolism. Moreover, RA pathobiology seems to share some common pathways with atherosclerosis, including endothelial dysfunction that is related to underlying chronic inflammation. The extent of the metabolic changes and the types of metabolites seen may be good markers of cytokine-mediated inflammatory processes in RA. Altered metabolic fingerprints may be useful in predicting the development of RA in patients with early arthritis as well as in the evaluation of the treatment response. Evidence supports the role of metabolomic analysis as a novel and nontargeted approach for identifying potential biomarkers and for improving the clinical and therapeutical management of patients with chronic inflammatory diseases. Here, we review the metabolic changes occurring in the pathogenesis of RA as well as the implication of the metabolic features in the treatment response.
Collapse
|
50
|
Chang KC. Cilostazol inhibits HMGB1 release in LPS-activated RAW 264.7 cells and increases the survival of septic mice. Thromb Res 2015; 136:456-64. [DOI: 10.1016/j.thromres.2015.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/11/2015] [Accepted: 06/14/2015] [Indexed: 10/23/2022]
|