1
|
Tang N, Luo X, Ding Z, Shi Y, Cao X, Wu S. Single-Cell Multi-Dimensional data analysis reveals the role of ARL4C in driving rheumatoid arthritis progression and Macrophage polarization dynamics. Int Immunopharmacol 2024; 141:112987. [PMID: 39182267 DOI: 10.1016/j.intimp.2024.112987] [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: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Rheumatoid arthritis (RA) is an enduring autoimmune inflammatory condition distinguished by continual joint inflammation, hyperplasia of the synovium, erosion of bone, and deterioration of cartilage.Fibroblast-like synoviocytes (FLSs) exhibiting "tumor-like" traits are central to this mechanism.ADP-ribosylation factor-like 4c (ARL4C) functions as a Ras-like small GTP-binding protein, significantly impacting tumor migration, invasion, and proliferation.However, it remains uncertain if ARL4C participates in the stimulation of RA FLSs exhibiting "tumor-like" features, thereby fostering the advancement of RA. In our investigation, we unveiled, for the inaugural instance, via the amalgamated scrutiny of single-cell RNA sequencing (scRNA-seq) and Bulk RNA sequencing (Bulk-seq) datasets, that activated fibroblast-like synoviocytes (FLSs) showcase high expression of ARL4C, and the ARL4C protein expression in FLSs derived from RA patients significantly surpasses that observed in individuals with osteoarthritis (OA) and traumatic injury (trauma).Silencing of the ARL4C gene markedly impeded the proliferation of RA FLSs by hindered the transition of cells from the G0/G1 phase to the S phase, and intensified cell apoptosis and diminished the migratory and invasive capabilities. Co-culture of ARL4C gene-silenced RA FLSs with monocytes/macrophages significantly inhibited the polarization of monocytes/macrophages toward M1 and the repolarization of M2 to M1.Furthermore, intra-articular injection of shARL4C significantly alleviated synovial inflammation and cartilage erosion in collagen-induced arthritis (CIA) rats. In conclusion, our discoveries propose that ARL4C assumes a central role in the synovial inflammation, cartilage degradation, and bone erosion associated with RA by triggering the PI3K/AKT and MAPK signaling pathways within RA FLSs.ARL4C holds promise as a prospective target for the development of pharmaceutical agents targeting FLSs, with the aim of addressing RA.
Collapse
Affiliation(s)
- Ning Tang
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xin Luo
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Zhiyu Ding
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yanbin Shi
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xu Cao
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Song Wu
- Department of Orthopaedics, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
2
|
Li R, Kuang Y, Niu Y, Zhang S, Chen S, Su F, Wang J, Lin S, Liu D, Shen C, Liang L, Zheng SG, Jie L, Xiao Y, Xu H. FTO-mediated RNA m 6A methylation regulates synovial aggression and inflammation in rheumatoid arthritis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167341. [PMID: 39025373 DOI: 10.1016/j.bbadis.2024.167341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Fibroblast-like synoviocytes (FLS) plays an important role in synovial inflammation and joint damage in rheumatoid arthritis (RA). As the most abundant mRNA modification, N6-methyladenosine (m6A) is involved in the development of various diseases; however, its role in RA remains to be defined. In this study, we reported the elevated expression of the m6A demethylase fat mass and obesity-associated protein (FTO) in FLS and synovium from RA patients. Functionally, FTO knockdown or treatment with FB23-2, an inhibitor of the mRNA m6A demethylase FTO, inhibited the migration, invasion and inflammatory response of RA FLS, however, FTO-overexpressed RA FLS exhibited increased migration, invasion and inflammatory response. We further demonstrated that FTO promoted ADAMTS15 mRNA stability in an m6A-IGF2BP1 dependent manner. Notably, the severity of arthritis was significantly reduced in CIA mice with FB23-2 administration or CIA rats with intra-articular injection of FTO shRNA. Our results illustrate the contribution of FTO-mediated m6A modification to joint damage and inflammation in RA and suggest that FTO might be a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Ruiru Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Yu Kuang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Yuanyuan Niu
- Department of General Practice, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Shuoyang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Simin Chen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Fan Su
- Department of Geriatrics, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Jingnan Wang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Shuibin Lin
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Di Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Chuyu Shen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Liuqin Liang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Song Jiang Research Institutes, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Ligang Jie
- Department of Rheumatology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Youjun Xiao
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China.
| | - Hanshi Xu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, China.
| |
Collapse
|
3
|
Liu S, Hall DJ, Dommann-Scherrer C, Pourzal R, Wahl P. Fourier-transform infrared spectroscopy imaging is a useful adjunct to routine histopathology to identify failure of polyethylene inlays in revision total hip arthroplasty. APMIS 2024; 132:553-563. [PMID: 38741279 DOI: 10.1111/apm.13421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
The use of highly crosslinked ultra-high molecular weight polyethylene (XLPE) has significantly reduced the volumetric wear of acetabular liners, thereby reducing the incidence of osteolysis. However, contemporary components tend to generate smaller wear particles, which can no longer be identified using conventional histology. This technical limitation can result in imprecise diagnosis. Here, we report on two uncemented total hip arthroplasty cases (~7 years in situ) revised for periprosthetic fracture of the femur and femoral loosening, respectively. Both liners exhibited prominent wear. The retrieved pseudocapsular tissue exhibited a strong macrophage infiltration without microscopically identifiable polyethylene particles. Yet, using Fourier-transform infrared micro-spectroscopic imaging (FTIR-I), we demonstrated the prominent intracellular accumulation of polyethylene debris in both cases. This study shows that particle induced osteolysis can still occur with XLPE liners, even under 10 years in situ. Furthermore, we demonstrate the difficulty of determining the presence of polyethylene debris within periprosthetic tissue. Considering the potentially increased bioactivity of finer particles from XLPE compared to conventional liners, an accurate detection method is required, and new histopathological hallmarks of particle induced osteolysis are needed. FTIR-I is a great tool to that end and can help the accurate determination of foreign body tissue responses.
Collapse
Affiliation(s)
- Songyun Liu
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Deborah J Hall
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | | | - Robin Pourzal
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Peter Wahl
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
- Faculty of Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Ou K, Zhang Q, Xi F, Ni H, Lu J, Lyu X, Wang C, Li Q, Wang Q. Prenatal EGCG consumption impacts hepatic glycogen synthesis and lipid metabolism in adult mice. Int J Biol Macromol 2024; 260:129491. [PMID: 38228202 DOI: 10.1016/j.ijbiomac.2024.129491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/27/2023] [Accepted: 01/12/2024] [Indexed: 01/18/2024]
Abstract
In this study, the impact of prenatal exposure to Epigallocatechin gallate (EGCG) on the liver of adult offspring mice was investigated. While EGCG is known for its health benefits, its effects of prenatal exposure on the liver remain unclear. Pregnant C57BL/6 J mice were exposed to 1 mg/kg of EGCG for 16 days to assess hepatotoxicity effects of adult offspring. Transcriptomics and metabolomics were employed to elucidate the hepatotoxicity mechanisms. The findings revealed that prenatal EGCG exposure led to a decrease in liver somatic index, enhanced inflammatory responses and disrupted liver function through increased glycogen accumulation in adult mice. The integrated omics analysis revealed significant alterations in key pathways involved in liver glucose lipid metabolism, such as gluconeogenesis, dysregulation of insulin signaling, and induction of liver inflammation. Furthermore, the study found a negative correlation between the promoter methylation levels of Ppara and their mRNA levels, suggesting that EGCG could reduce hepatic lipid content through epigenetic modifications. The findings suggest that prenatal EGCG exposure can have detrimental impacts on the liver among adult individuals and emphasize the need for a comprehensive evaluation of the potential risks associated with EGCG consumption during pregnancy.
Collapse
Affiliation(s)
- Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Quan Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Feifei Xi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Huizhen Ni
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Jiebo Lu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Xuejing Lyu
- School of Medicine, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Qiyuan Li
- School of Medicine, Xiamen University, Xiamen, Fujian 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, China.
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China.
| |
Collapse
|
5
|
Kuang Y, Li R, Wang J, Xu S, Qiu Q, Lin S, Liu D, Shen C, Liu Y, Xu M, Lin W, Zhang S, Liang L, Xu H, Xiao Y. ALKBH5-Mediated RNA m 6 A Methylation Regulates the Migration, Invasion, and Proliferation of Rheumatoid Fibroblast-Like Synoviocytes. Arthritis Rheumatol 2024; 76:192-205. [PMID: 37584615 DOI: 10.1002/art.42676] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 06/27/2023] [Accepted: 08/09/2023] [Indexed: 08/17/2023]
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLSs) are critical for promoting joint damage in rheumatoid arthritis (RA). N6 -methyladenosine (m6 A) modification plays key roles in various diseases, but its role in the pathogenesis of RA is largely unknown. Here, we investigate increased demethylase ALKBH5 promotion of proliferation, migration, and invasion of RA FLSs via regulating JARID2 expression. METHODS ALKBH5 expression in FLSs was evaluated using real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot. 5-ethynyl-2'-deoxyuridine, scratch wound healing, and transwell assays were implemented to determine the role of ALKBH5 on RA FLS proliferation, mobility, and migration. Then, m6 A sequencing combined with RNA sequencing was performed to identify the potential targets of ALKBH5. RNA immunoprecipitation and RNA pulldown were then used to validate the interaction between the protein and messenger RNA (mRNA). Collagen-induced arthritis (CIA) and delayed-type hypersensitivity arthritis (DTHA) models were further established to assess the therapeutic potency of ALKBH5 in vivo. RESULTS We demonstrated that ALKBH5 expression was increased in FLSs and synovium from RA. Functionally, ALKBH5 knockdown inhibited the proliferation, migration, and invasion of RA FLSs, whereas overexpression of ALKBH5 displayed the opposite effect. Mechanistically, ALKBH5 mediated m6 A modification in the JARID2 mRNA and enhanced its mRNA stability in cooperation with IGF2BP3. Intriguingly, the severity of arthritis was attenuated in mice with DTHA and ALKBH5 knockout or rats with CIA and intra-articular injection of ALKBH5 short hairpin RNA. CONCLUSION Our findings suggest that ALKBH5-mediated m6 A modification is crucial for synovial hyperplasia and invasion in RA. ALKBH5 might be a potential therapeutic target for RA and even for dysregulated fibroblasts in a wide range of diseases.
Collapse
Affiliation(s)
- Yu Kuang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ruiru Li
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jingnan Wang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Siqi Xu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qian Qiu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shuibin Lin
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Di Liu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chuyu Shen
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yingli Liu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meilin Xu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Lin
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shuoyang Zhang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liuqin Liang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hanshi Xu
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Youjun Xiao
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
6
|
Straub RH, Pongratz G, Buttgereit F, Gaber T. [Energy metabolism of the immune system : Consequences in chronic inflammation]. Z Rheumatol 2023:10.1007/s00393-023-01389-4. [PMID: 37488246 DOI: 10.1007/s00393-023-01389-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Energy is the currency of life. The systemic and intracellular energy metabolism plays an essential role for the energy supply of the resting and activated immune system and this also applies to chronic inflammatory diseases. OBJECTIVE This presentation examines both components of the systemic and cellular energy metabolism in health and chronic inflammation. MATERIAL AND METHODS A literature search was conducted using PubMed, Embase and the Cochrane Library. The information is presented in the form of a narrative review. RESULTS A chronically activated immune system acquires large amounts of energy-rich substrates that are lost for other functions of the body. In particular, the immune system and the brain are in competition. The consequences of this competition are many known diseases, such as fatigue, anxiety, depression, anorexia, sleep problems, sarcopenia, osteoporosis, insulin resistance, hypertension and others. The permanent change in the brain causes long-term alterations that stimulate disease sequelae even after disease remission. In the intracellular energy supply, chronic inflammation typically involves a conversion to glycolysis (to lactate, which has its own regulatory functions) and the pentose phosphate pathway in disorders of mitochondrial function. The chronic changes in immune cells of patients with rheumatoid arthritis (RA) lead to a disruption of the citric acid cycle (Krebs cycle). The hypoxic situation in the inflamed tissue stimulates many alterations. A differentiation is made between effector functions and regulatory functions of immune cells. CONCLUSION Based on the energy changes mentioned, novel treatment suggestions can be made in addition to those already known in energy metabolism.
Collapse
Affiliation(s)
- Rainer H Straub
- Labor für Experimentelle Rheumatologie und Neuroendokrin-Immunologie, Klinik und Poliklinik für Innere Medizin I, Universitätsklinikum Regensburg, 93042, Regensburg, Deutschland.
| | - Georg Pongratz
- Abteilung für Rheumatologie, Klinik für Gastroenterologie, Krankenhaus Barmherzige Brüder Regensburg, 93049, Regensburg, Deutschland
| | - Frank Buttgereit
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Deutschland
| | - Timo Gaber
- Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Deutschland
| |
Collapse
|
7
|
Pucino V, Nefla M, Gauthier V, Alsaleh G, Clayton SA, Marshall J, Filer A, Clark AR, Raza K, Buckley CD. Differential effect of lactate on synovial fibroblast and macrophage effector functions. Front Immunol 2023; 14:1183825. [PMID: 37304267 PMCID: PMC10251493 DOI: 10.3389/fimmu.2023.1183825] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction The synovial membrane is the main site of inflammation in rheumatoid arthritis (RA). Here several subsets of fibroblasts and macrophages, with distinct effector functions, have been recently identified. The RA synovium is hypoxic and acidic, with increased levels of lactate as a result of inflammation. We investigated how lactate regulates fibroblast and macrophage movement, IL-6 secretion and metabolism via specific lactate transporters. Methods Synovial tissues were taken from patients undergoing joint replacement surgery and fulfilling the 2010 ACR/EULAR RA criteria. Patients with no evidence of degenerative or inflammatory disease were used as control. Expression of the lactate transporters SLC16A1 and SLC16A3 on fibroblasts and macrophages was assessed by immunofluorescence staining and confocal microscopy. To test the effect of lactate in vitro we used RA synovial fibroblasts and monocyte-derived macrophages. Migration was assessed via scratch test assays or using trans-well inserts. Metabolic pathways were analysed by Seahorse analyser. IL-6 secretion was determined by ELISA. Bioinformatic analysis was performed on publicly available single cell and bulk RNA sequencing datasets. Results We show that: i) SLC16A1 and SLC16A3 which regulate lactate intake and export respectively, are both expressed in RA synovial tissue and are upregulated upon inflammation. SLC16A3 is more highly expressed by macrophages, while SLC16A1 was expressed by both cell types. ii) This expression is maintained in distinct synovial compartments at mRNA and protein level. iii) Lactate, at the concentration found in RA joints (10 mM), has opposite effects on the effector functions of these two cell types. In fibroblasts, lactate promotes cell migration, IL-6 production and increases glycolysis. In contrast macrophages respond to increases in lactate by reducing glycolysis, migration, and IL-6 secretion. Discussion In this study, we provide the first evidence of distinct functions of fibroblasts and macrophages in presence of high lactate levels, opening new insights in understanding the pathogenesis of RA and offering novel potential therapeutic targets.
Collapse
Affiliation(s)
- Valentina Pucino
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Kennedy Institute of Rheumatology, Oxford University, Oxford, United Kingdom
| | - Meriam Nefla
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Vincent Gauthier
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Kennedy Institute of Rheumatology, Oxford University, Oxford, United Kingdom
| | - Ghada Alsaleh
- Kennedy Institute of Rheumatology, Oxford University, Oxford, United Kingdom
| | - Sally A. Clayton
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Jennifer Marshall
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Andrew Filer
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Andy R. Clark
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Karim Raza
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Sandwell and West Birmingham National Health System (NHS) Trust, Birmingham, United Kingdom
| | - Christopher D. Buckley
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- Kennedy Institute of Rheumatology, Oxford University, Oxford, United Kingdom
| |
Collapse
|
8
|
Jing W, Liu C, Su C, Liu L, Chen P, Li X, Zhang X, Yuan B, Wang H, Du X. Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Front Immunol 2023; 14:1107670. [PMID: 36845127 PMCID: PMC9948260 DOI: 10.3389/fimmu.2023.1107670] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, pannus formation, and bone and cartilage damage. It has a high disability rate. The hypoxic microenvironment of RA joints can cause reactive oxygen species (ROS) accumulation and mitochondrial damage, which not only affect the metabolic processes of immune cells and pathological changes in fibroblastic synovial cells but also upregulate the expression of several inflammatory pathways, ultimately promoting inflammation. Additionally, ROS and mitochondrial damage are involved in angiogenesis and bone destruction, thereby accelerating RA progression. In this review, we highlighted the effects of ROS accumulation and mitochondrial damage on inflammatory response, angiogenesis, bone and cartilage damage in RA. Additionally, we summarized therapies that target ROS or mitochondria to relieve RA symptoms and discuss the gaps in research and existing controversies, hoping to provide new ideas for research in this area and insights for targeted drug development in RA.
Collapse
Affiliation(s)
- Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cui Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chenghong Su
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Limei Liu
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ping Chen
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangjun Li
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xinghua Zhang
- Department of Acupuncture, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bo Yuan
- Department of Acupuncture and Pain, Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Haidong Wang
- Department of Rheumatic and Bone Disease, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaozheng Du
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
9
|
Cao L, Zheng X, Han P, Ren L, Hu F, Li Z. Raman spectroscopy as a promising diagnostic method for rheumatoid arthritis. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:709-718. [PMID: 36598183 DOI: 10.1039/d2ay01904c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Background: Diagnosis of rheumatoid arthritis (RA) basically relies on clinical symptoms and autoantibodies, especially anti-citrullinated protein antibodies (ACPAs) and rheumatoid factor (RF). However, the lack of autoantibodies is still a dilemma clinically in seronegative RA, especially in the early stage of the disease. This study aimed to provide a unique disease fingerprint with high diagnostic value to discriminate RA based on Raman spectroscopy. Methods: Raman spectroscopy provides a repertoire of biomolecules in serum from RA. Multivariate dimension-reducing methods and machine-learning algorithms were exploited to reveal the intrinsic differences and the potential discrimination power. The underlying differential biomolecules were retrieved by the assignment of Raman peaks. Moreover, the correlations between the spectral differences and RA patient's clinical and immunological manifestations were also analyzed. Results: RA patients exhibited unique Raman spectra characterized by biomolecular alterations during the disease progression. The discrimination power yielded 97.3% sensitivity and 94.8% specificity for RA diagnosis. In the recognition of ACPA-negative RA, the sensitivity and specificity also reached 95.6% and 92.8%, respectively. In particular, the differential Raman spectrum peaks of RA patients mainly represented lipids, amino acids, glycogen, and fatty acids. Further analysis showed that the different serum Raman spectra correlated with the clinical features of RA, including disease duration, RF, anticyclic citrullinated peptide antibodies (anti-CCPs), IgA, IgM, IgG, tender joint count, and swollen joint count (|rs| = 0.15-0.52, p < 0.05). Conclusions: Raman spectroscopy was revealed to be a promising diagnostic method for RA, especially for ACPA-negative patients.
Collapse
Affiliation(s)
- Lulu Cao
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Xi Zheng
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Peng Han
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Limin Ren
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital and Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
10
|
Cheng DW, Yue YF, Chen CX, Hu YD, Tang Q, Xie M, Liu L, Li D, Zhu HL, Cheng ML. Emodin alleviates arthritis pain through reducing spinal inflammation and oxidative stress. Mol Pain 2022; 18:17448069221146398. [PMID: 36474308 PMCID: PMC9772972 DOI: 10.1177/17448069221146398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic pain is the predominant problem for rheumatoid arthritis patients, and negatively affects quality of life. Arthritis pain management remains largely inadequate, and developing new treatment strategies are urgently needed. Spinal inflammation and oxidative stress contribute to arthritis pain and represent ideal targets for the treatment of arthritis pain. In the present study, collagen-induced arthritis (CIA) mouse model was established by intradermally injection of type II collagen (CII) in complete Freund's adjuvant (CFA) solution, and exhibited as paw and ankle swelling, pain hypersensitivity and motor disability. In spinal cord, CIA inducement triggered spinal inflammatory reaction presenting with inflammatory cells infiltration, increased Interleukin-1β (IL-1β) expression, and up-regulated NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and cleaved caspase-1 levels, elevated spinal oxidative level presenting as decreased nuclear factor E2-related factor 2 (Nrf2) expression and Superoxide dismutase (SOD) activity. To explore potential therapeutic options for arthritis pain, emodin was intraperitoneally injected for 3 days on CIA mice. Emodin treatment statistically elevated mechanical pain sensitivity, suppressed spontaneous pain, recovered motor coordination, decreased spinal inflammation score and IL-1β expression, increased spinal Nrf2 expression and SOD activity. Further, AutoDock data showed that emodin bind to Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) through two electrovalent bonds. And emodin treatment increased the phosphorylated AMPK at threonine 172. In summary, emodin treatment activates AMPK, suppresses NLRP3 inflammasome response, elevates antioxidant response, inhibits spinal inflammatory reaction and alleviates arthritis pain.
Collapse
Affiliation(s)
- Ding-Wen Cheng
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yuan-Fen Yue
- Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Chun-Xi Chen
- Xishui Affiliated Hospital of Hubei University of Science and Technology, Huanggang, China
| | - Yin-Di Hu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qiong Tang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Min Xie
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ling Liu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Dai Li
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hai-Li Zhu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China,Hai-Li Zhu, Xianning Medical College, Hubei University of Science and Technology, No. 88 Xianning Road, Xianning, Hubei 437100, China.
| | - Meng-Lin Cheng
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China,Meng-Lin Cheng, Xianning Medical College, Hubei University of Science and Technology, No. 88 Xianning Road, Xianning, Hubei 437100, China.
| |
Collapse
|
11
|
Terrón-Puig M, Huber-Ruano I, Sabadell-Basallote J, Ejarque M, Núñez-Roa C, Maymó-Masip E, Jorba R, Serena C, Vendrell J, Fernández-Veledo S. Glycogen accumulation in adipocyte precursors from elderly and obese subjects triggers inflammation via SIRT1/6 signaling. Aging Cell 2022; 21:e13667. [PMID: 35811457 PMCID: PMC9381900 DOI: 10.1111/acel.13667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/18/2022] [Accepted: 06/24/2022] [Indexed: 12/13/2022] Open
Abstract
Dysfunctional adipocyte precursors have emerged as key determinants for obesity- and aging-related inflammation, but the mechanistic basis remains poorly understood. Here, we explored the dysfunctional adipose tissue of elderly and obese individuals focusing on the metabolic and inflammatory state of human adipose-derived mesenchymal stromal cells (hASCs), and on sirtuins, which link metabolism and inflammation. Both obesity and aging impaired the differentiation potential of hASCs but had a different impact on their proliferative capacity. hASCs from elderly individuals (≥65 years) showed an upregulation of glycolysis-related genes, which was accompanied by increased lactate secretion and glycogen storage, a phenotype that was exaggerated by obesity. Multiplex protein profiling revealed that the metabolic switch to glycogenesis was associated with a pro-inflammatory secretome concomitant with a decrease in the protein expression of SIRT1 and SIRT6. siRNA-mediated knockdown of SIRT1 and SIRT6 in hASCs from lean adults increased the expression of pro-inflammatory and glycolysis-related markers, and enforced glycogen deposition by overexpression of protein targeting to glycogen (PTG) led to a downregulation of SIRT1/6 protein levels, mimicking the inflammatory state of hASCs from elderly subjects. Overall, our data point to a glycogen-SIRT1/6 signaling axis as a driver of age-related inflammation in adipocyte precursors.
Collapse
Affiliation(s)
- Margarida Terrón-Puig
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Rovira i Virgili University, Tarragona, Spain.,Servei d'Anàlisis clíniques. Laboratori Clínic ICS Camp de Tarragona-Terres de l'Ebre, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Isabel Huber-Ruano
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joan Sabadell-Basallote
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Rovira i Virgili University, Tarragona, Spain
| | - Miriam Ejarque
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Catalina Núñez-Roa
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Elsa Maymó-Masip
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosa Jorba
- Servei de Cirugia General i de l'Aparell Digestiu, Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Carolina Serena
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Joan Vendrell
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Rovira i Virgili University, Tarragona, Spain
| | - Sonia Fernández-Veledo
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Endocrinology and Nutrition Service, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
12
|
Fusco C, Nardella G, Di Filippo L, Dejana E, Cacchiarelli D, Petracca A, Micale L, Malinverno M, Castori M. Transcriptome Analysis Reveals Altered Expression of Genes Involved in Hypoxia, Inflammation and Immune Regulation in Pdcd10-Depleted Mouse Endothelial Cells. Genes (Basel) 2022; 13:genes13060961. [PMID: 35741725 PMCID: PMC9222422 DOI: 10.3390/genes13060961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebral cavernous malformations (CCM) are capillary malformations affecting the central nervous system and commonly present with headaches, epilepsy and stroke. Treatment of CCM is symptomatic, and its prevention is limited. CCM are often sporadic but sometimes may be multifocal and/or affect multiple family members. Heterozygous pathogenic variants in PDCD10 cause the rarest and apparently most severe genetic variant of familial CCM. We carried out an RNA-Seq and a Q-PCR validation analysis in Pdcd10-silenced and wild-type mouse endothelial cells in order to better elucidate CCM molecular pathogenesis. Ninety-four differentially expressed genes presented an FDR-corrected p-value < 0.05. A functionally clustered dendrogram showed that differentially expressed genes cluster in cell proliferation, oxidative stress, vascular processes and immune response gene-ontology functions. Among differentially expressed genes, the major cluster fell in signaling related to inflammation and pathogen recognition, including HIF1α and Nos2 signaling and immune regulation. Validation analysis performed on wild-type, Pdcd10-null and Pdcd10-null reconstituted cell lines was consistent with RNA-Seq data. This work confirmed previous mouse transcriptomic data in endothelial cells, which are recognized as a critical tissue for CCM formation and expands the potential molecular signatures of PDCD10-related familial CCM to alterations in inflammation and pathogen recognition pathways.
Collapse
Affiliation(s)
- Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 Foggia, Italy; (G.N.); (A.P.); (L.M.); (M.C.)
- Correspondence: ; Tel.: +39-0882-416350; Fax: +39-0882-411616
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 Foggia, Italy; (G.N.); (A.P.); (L.M.); (M.C.)
| | | | - Elisabetta Dejana
- Vascular Biology Unit, FIRC Institute of Molecular Oncology Foundation (IFOM), 20139 Milan, Italy; (E.D.); (M.M.)
| | - Davide Cacchiarelli
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy;
- Department of Translational Medicine, University of Naples “Federico II”, 80126 Naples, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples “Federico II”, 80126 Naples, Italy
| | - Antonio Petracca
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 Foggia, Italy; (G.N.); (A.P.); (L.M.); (M.C.)
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 Foggia, Italy; (G.N.); (A.P.); (L.M.); (M.C.)
| | - Matteo Malinverno
- Vascular Biology Unit, FIRC Institute of Molecular Oncology Foundation (IFOM), 20139 Milan, Italy; (E.D.); (M.M.)
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 Foggia, Italy; (G.N.); (A.P.); (L.M.); (M.C.)
| |
Collapse
|
13
|
Liu J, Xue J, Xu B, Yu J, Zhang Y, Qin L, Zhang A, Yang Y. miR-135a-5p mediated down-regulation of STAT6 inhibits proliferation and induces apoptosis of fibroblast-like synoviocytes in rheumatoid arthritis. Am J Transl Res 2022; 14:3092-3103. [PMID: 35702088 PMCID: PMC9185078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/24/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Rheumatoid arthritis (RA), as a chronic autoimmune disorder, seriously threatens human health. However, no study has thoroughly illustrated the etiology of RA. The present work focused on investigating the biological functions of STAT6 and the upstream miRNAs that regulate its expression. METHODS Synovial tissues from rheumatoid arthritis (RA) patients and normal participants were acquired. Cell viability, proliferation, apoptosis, concentrations of cytokines, miRNA and protein levels, and relative luciferase activities were detected. RESULTS WB and qRT-PCR showed that STAT6 was obviously up-regulated in synovial tissues of RA patients as well as RA fibroblast-like synoviocytes (RA FLSs). Functionally, down-regulation of STAT6 significantly inhibited the growth of RA FLSs as indicated by EdU and CCK-8 assays. In addition, inhibition of STAT6 remarkably promoted apoptosis of RA FLSs. Besides, silence of STAT6 notably suppressed inflammatory cytokine levels, such as TNF-α, IL-6 and IL-1β. Mechanistically, STAT6 was predicted to be the direct target of and negatively regulated by miR-135a-5p. Moreover, STAT6 was involved in the regulation of miR-135a-5p on cell growth, apoptosis and inflammatory response of RA FLSs. CONCLUSION miR-135a-5p/STAT6 is a potential novel therapeutic target for RA treatment.
Collapse
Affiliation(s)
- Jiajia Liu
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Jianhua Xue
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Bo Xu
- Department of Orthopedics, Qidong Hospital of Traditional Chinese MedicineNantong, Jiangsu Province, China
| | - Junbo Yu
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yuxuan Zhang
- Department of Foot and Ankle Surgery, Wuxi No. 9 People’s Hospital Affiliated to Soochow UniversityWuxi, Jiangsu Province, China
| | - Lili Qin
- Department of Endoscopic Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Aixian Zhang
- Department of General Practice Medicine, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yang Yang
- Department of Trauma Center, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| |
Collapse
|
14
|
Cao J, Ni Y, Zhang H, Ning X, Qi X. Inhibition of Kruppel-like factor 7 attenuates cell proliferation and inflammation of fibroblast-like synoviocytes in rheumatoid arthritis through NF-κB and MAPK signaling pathway. Exp Anim 2022; 71:356-367. [PMID: 35321971 PMCID: PMC9388335 DOI: 10.1538/expanim.21-0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease, which can lead to joint inflammation and progressive joint destruction. Kruppel-like factor 7 (KLF7) is the member of KLF family and
plays an important role in multiple biological progresses. However, its precise roles in RA have not been described. Present study aimed to investigate the role of KLF7 in RA-fibroblast-like
synoviocytes (FLSs). Data showed that KLF7 expression was obviously upregulated in synovial tissues of rats with adjuvant-induced arthritis. Functional studies demonstrated that the loss of
KLF7 may suppress cell proliferation and the expression of pro-inflammatory factors (IL-6, IL-1β, IL-17A) and matrix metalloproteinase (MMP-1, MMP-3, MMP-13) in FLSs through the inhibition
of phosphorylation of nuclear factor κB (NF-κB) p65 and JNK. We further showed that miR-9a-5p specifically interacts with KLF7 to negatively regulate the expression of KLF7 in RA-FLSs. Taken
together, our results demonstrated that KLF7 which targeted by miR-9a-5p might participate in the pathogenesis of RA by promoting cell proliferation, pro-inflammatory cytokine release and
MMP expression through the activation of NF-κB and JNK pathways in RA-FLSs. Hence, KLF7 could be a novel target for RA therapy.
Collapse
Affiliation(s)
- Jingjing Cao
- Teaching and Research Section of Internal Medicine, Hebei Medical University.,Department of Rheumatology and Immunology, Hebei General Hospital
| | - Yanhui Ni
- Department of Cardiology, Hebei General Hospital
| | | | - Xiaoran Ning
- Department of Rheumatology and Immunology, Hebei General Hospital
| | - Xiaoyong Qi
- Teaching and Research Section of Internal Medicine, Hebei Medical University.,Department of Cardiology Center, Hebei General Hospital
| |
Collapse
|
15
|
Chen D, Cai X, Ouyang H, Yuan S, Wang X, Lin L, Chen Z, Huang M. Increased eEF2K Promotes Glycolysis and Aggressive Behaviors of Fibroblast-Like Synoviocytes in Rheumatoid Arthritis. J Inflamm Res 2022; 15:1729-1744. [PMID: 35300214 PMCID: PMC8922331 DOI: 10.2147/jir.s337620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/01/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Aggressive phenotype and abnormal glycolytic metabolism of fibroblast-like synoviocytes (FLSs) are essential to joint inflammation and damage in rheumatoid arthritis (RA). Eukaryotic elongation factor-2 kinase (eEF2K) is a negative regulator of protein synthesis and has been shown to play an important role in regulating various cellular processes and promoting glycolysis in tumor cells. However, the role of eEF2K in regulating the pathogenic FLS behaviors is unknown. Methods A specific inhibitor of eEF2K, NH125, and siRNA were used to evaluate the role of eEF2K on RA FLSs in vitro. Collagen-induced arthritis (CIA) mice were used to evaluate the in vivo effect of eEF2K. Cell migration, invasion of RA FLSs were assessed by transwell or wound healing assays. Relative changes of cytokines were analyzed by quantitative real-time PCR, western blot and ELISA. Results Herein, we found an increased expression of eEF2K in synovial tissues and FLSs of RA patients. eEF2K knockdown by siRNA or treatment with NH125, an inhibitor of eEF2K, significantly reduced inflammation, migration/invasion, glucose uptake and lactate productions. eEF2K knockdown suppressed TNF-α-induced activation of NF-κB and AKT pathways in RA FLSs. Lactate reversed the inhibitory effect of eEF2K knockdown on inflammation and migration of RA FLSs. Moreover, lactate was also involved in eEF2K-mediated activation of NF-κB and AKT. NH125 treatment attenuated the severity of arthritis in collagen-induced arthritis mice. Conclusion eEF2K inhibition suppressed glycolysis and aggressive behaviors of RA FLS, which indicated that targeting eEF2K may be a new strategy for the treatment of RA.
Collapse
Affiliation(s)
- Dongying Chen
- Department of Rheumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guandong, People’s Republic of China
| | - Xiaoyan Cai
- Department of Rheumatology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guandong, People’s Republic of China
| | - Hui Ouyang
- Department of Digestive Medicine Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, ShenZhen, Guandong, People’s Republic of China
| | - Shiwen Yuan
- Department of Rheumatology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guandong, People’s Republic of China
| | - Xiaodong Wang
- Department of Ultrasound, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guandong, People’s Republic of China
| | - Lian Lin
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People’s Republic of China
| | - Zhiqing Chen
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People’s Republic of China
| | - Mingcheng Huang
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People’s Republic of China
- Correspondence: Mingcheng Huang, Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People’s Republic of China, Email
| |
Collapse
|
16
|
Liu K. Immune, metabolism and therapeutic targets in RA (Rheumatoid Arthritis). BIO WEB OF CONFERENCES 2022. [DOI: 10.1051/bioconf/20225501016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Rheumatoid arthritis is a classic autoimmune disease, the pathogenesis of which is closely linked to the auto-reactivity of immune cells and joint inflammation. Three cell types, namely T cells, macrophages and fibroblast-like synoviocytes (FLS), play an important role in the pathogenesis of RA. Numerous studies have pointed to a metabolic reprogramming of T cells, macrophages and FLS in the pathogenesis of RA arthritis, with alterations in different metabolic pathways of cells, mainly producing a shift from oxidative phosphorylation (OXPHOS) to glycolysis, in addition to lipid metabolism and amino acid metabolism which are also altered in the cellular activation state. Metabolic changes are regulated by metabolism-related signalling pathways, and RA is associated with two representative signalling pathways, namely the mTOR signalling pathway and the AMPK signalling pathway. In RA, both signalling pathways are activated or inhibited, and through a series of cascade reactions, different gene expressions are ultimately induced, altering intracellular metabolic pathways and promoting pro-inflammatory functions (e.g. pro-inflammatory cytokine release and FLS phenotypes), or inhibiting the expression of genes related to immune tolerance. Targeting key components of metabolic signalling pathways and key enzymes in cellular metabolic pathways in RA has emerged as a new way of finding drugs for RA, and many modulators targeting these targets have been extensively studied for their therapeutic effects in RA. In this article, we focus on cellular metabolic alterations in RA, related signalling pathways and possible drugs targeting RA metabolic pathways.
Collapse
|
17
|
Hanlon MM, Canavan M, Barker BE, Fearon U. Metabolites as drivers and targets in Rheumatoid Arthritis. Clin Exp Immunol 2021; 208:167-180. [PMID: 35020864 PMCID: PMC9188347 DOI: 10.1093/cei/uxab021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/03/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by neovascularization, immune cell infiltration, and synovial hyperplasia, which leads to degradation of articular cartilage and bone, and subsequent functional disability. Dysregulated angiogenesis, synovial hypoxia, and immune cell infiltration result in a ‘bioenergetic crisis’ in the inflamed joint which further exacerbates synovial invasiveness. Several studies have examined this vicious cycle between metabolism, immunity, and inflammation and the role metabolites play in these interactions. To add to this complexity, the inflamed synovium is a multicellular tissue with many cellular subsets having different metabolic requirements. Metabolites can shape the inflammatory phenotype of immune cell subsets during disease and act as central signalling hubs. In the RA joint, the increased energy demand of stromal and immune cells leads to the accumulation of metabolites such as lactate, citrate, and succinate as well as adipocytokines which can regulate downstream signalling pathways. Transcription factors such as HIF1ɑ and mTOR can act as metabolic sensors to activate synovial cells and drive pro-inflammatory effector function, thus perpetuating chronic inflammation further. These metabolic intermediates may be potential therapeutic targets and so understanding the complex interplay between metabolites and synovial cells in RA may allow for identification of novel therapeutic strategies but also may provide significant insight into the underlying mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Megan M Hanlon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin.,EULAR Centre of Excellence for Rheumatology, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin.,EULAR Centre of Excellence for Rheumatology, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, Dublin, Ireland
| | - Brianne E Barker
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin.,EULAR Centre of Excellence for Rheumatology, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin.,EULAR Centre of Excellence for Rheumatology, Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
18
|
Wei J, Fan S, Yu H, Shu L, Li Y. A New Strategy for the Rapid Identification and Validation of the Direct Targets of Aconitine-Induced Cardiotoxicity. Drug Des Devel Ther 2021; 15:4649-4664. [PMID: 34803375 PMCID: PMC8599306 DOI: 10.2147/dddt.s335461] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/28/2021] [Indexed: 12/22/2022] Open
Abstract
Background The interaction of small molecules with direct targets constitutes the molecular initiation events of drug efficacy and toxicity. Aconitine, an active compound of the Aconitum species, has various pharmacological effects but is strongly toxic to the heart. The direct targets of aconitine-induced cardiotoxicity remain unclear. Methods We predicted the toxic targets of aconitine based on network pharmacology and followed a novel proteomic approach based on the "drug affinity responsive target stability" technology combined with LC-MS/MS to identify the direct targets of aconitine. The identified targets were analysed from the perspective of multilevel and multidimensional bioinformatics through a network integration method. The binding sites were investigated via molecular docking to explore the toxicity mechanism and predict the direct targets of aconitine. Finally, atomic force microscopy (AFM) imaging was performed to verify the affinity of aconitine to the direct targets. Results PTGS2, predicted by network pharmacology as a toxic target, encodes cyclooxygenase 2 (COX-2), which is closely related to myocardial injury. Furthermore, cytosolic phospholipase A2 (cPLA2) is the upstream signal protein of PTGS2, and it is a key enzyme in the metabolism of arachidonic acid during an inflammatory response. We determined cPLA2 as a direct target, and AFM imaging verified that aconitine could bind to cPLA2 well; thus, aconitine may cause the expression of PTGS2/COX-2 and release inflammatory factors, thereby promoting myocardial injury and dysfunction. Conclusion We developed a complete set of methods to predict and verify the direct targets of aconitine, and cPLA2 was identified as one. Overall, the novel strategy provides new insights into the discovery of direct targets and the molecular mechanism of toxic components that are found in traditional Chinese medicine.
Collapse
Affiliation(s)
- Jinxia Wei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Simiao Fan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Hongxin Yu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Lexin Shu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
19
|
Huang M, Wu K, Zeng S, Liu W, Cui T, Chen Z, Lin L, Chen D, Ouyang H. Punicalagin Inhibited Inflammation and Migration of Fibroblast-Like Synoviocytes Through NF-κB Pathway in the Experimental Study of Rheumatoid Arthritis. J Inflamm Res 2021; 14:1901-1913. [PMID: 34012288 PMCID: PMC8126973 DOI: 10.2147/jir.s302929] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 12/26/2022] Open
Abstract
Background The aggressive phenotype of fibroblast-like synoviocytes (FLSs) is essential in the synovitis and bone destruction in rheumatoid arthritis (RA). Punicalagin is a natural polyphenol extracted in pomegranate juice, which possesses antioxidant, anti-inflammatory and anti-tumor properties suggesting it may be a potent drug for RA therapy. However, there is paucity of information on its effect in RA. Objective To investigate the effects of punicalagin on synovial inflammation and bone destruction in RA. Methods FLSs were isolated from synovial tissue of RA patients. The mRNA levels were evaluated by quantitative real-time PCR. Western blot was used for protein level measurements. The secretion of pro-inflammatory cytokines and metalloproteinases (MMPs) was detected by ELISA assays. Edu staining assays were carried out to investigate the proliferation of FLSs. Cell migration was assessed by Boyden chambers, wound scratch assays and F-actin staining in vitro. The intracellular translocation of nuclear factor kappa B (NF-κB) was investigated using immunofluorescence. The effects of punicalagin in vivo were measured by using collagen-induced arthritis (CIA) mice. Results Punicalagin treatments significantly reduced the TNF-α induced expressions of pro-inflammatory cytokines (IL-1β, IL-6, IL-8 and IL-17A) and MMPs (MMP-1 and MMP-13) of RA FLSs. Punicalagin also suppressed the proliferation and migration of RA FLSs. Moreover, punicalagin (50mg/kg/d) alleviated arthritis severity and bone destruction, and decreased serum IL-6 and TNF-α in CIA mice. Further mechanism studies indicated that punicalagin blocked NF-κB activation via suppressing phosphorylation of IKK and IkBα, and preventing the translocation of 65. Conclusion Our findings suggested that punicalagin might be one of natural therapeutic compounds for relieving RA progress via suppressing FLSs inflammation and migration through modulating NF-κB pathways.
Collapse
Affiliation(s)
- Mingcheng Huang
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| | - Keping Wu
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| | - Shan Zeng
- Department of Rheumatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guandong, People's Republic of China
| | - Wenfen Liu
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| | - Tianjiao Cui
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| | - Zhiqing Chen
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| | - Lian Lin
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| | - Dongying Chen
- Department of Rheumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guandong, People's Republic of China
| | - Hui Ouyang
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, ShenZhen, Guandong, People's Republic of China
| |
Collapse
|
20
|
Liu C, Zhao Q, Zhong L, Li Q, Li R, Li S, Li Y, Li N, Su J, Dhondrup W, Meng X, Zhang Y, Tu Y, Wang X. Tibetan medicine Ershiwuwei Lvxue Pill attenuates collagen-induced arthritis via inhibition of JAK2/STAT3 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113820. [PMID: 33465441 DOI: 10.1016/j.jep.2021.113820] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ershiwuwei Lvxue Pill (ELP, མགྲིན་མཚལ་ཉེར་ལྔ།), a traditional Tibetan medicine preparation, has been used hundreds of years for the clinical treatment of rheumatoid arthritis (RA) in the highland region of Tibet, China. However, the underlying mechanism of its therapeutic effect remains unclear. AIM OF THE STUDY The present study aimed to investigate the potential pharmacological mechanisms of anti-arthritic effect of ELP. MATERIALS AND METHODS The main chemical constituents of ELP were analyzed by ultra-performance liquid chromatography quadrupole-time-flight mass spectrometry (UPLC-Q-TOF-MS). Forty-eight male Wistar rats (220 ± 20 g) were randomly divided into six groups: normal group, collagen-induced arthritis (CIA) group, methotrexate group (1.05 mg/kg), ELP groups (115, 230 and 460 mg/kg). CIA rat models were assigned to evaluate the anti-RA activity of ELP by determining the paws swelling, arthritis score, organ coefficients of spleen and thymus, and histopathological analysis of knee joints of synovial tissues. The levels of TNF-α, IL-10, IL-6 and IL-17 in serum were measured by ELISA. In addition, mRNA and protein expression levels associated with JAK2/STAT3 signaling pathway in synovial tissues of CIA rats were detected by qRT-PCR, immunohistochemistry and Western blot analyses. RESULTS Fourteen main chemical constituents of ELP were quantitatively determined by UPLC-Q-TOF-MS analysis. Treatment with ELP reduced the paw swelling, arthritis score and organ coefficients of spleen and thymus. Histopathological examination revealed the protective effects of ELP on CIA rats with knee joint injury. The levels of serum pro-inflammatory cytokines (TNF-α, IL-6 and IL-17) were markedly reduced while the anti-inflammatory cytokine IL-10 was significantly increased with the treatment of ELP. Further investigations showed ELP down-regulated the mRNA and protein expression levels of Bcl-2, whereas up-regulated Bax, SOCS1 and SOCS3. Meanwhile, the ratios of p-JAK2/JAK2 and p-STAT3/STAT3 proteins from synovial tissues were dramatically decreased with the treatment of ELP, whereas no changes of the mRNA and protein expression levels of JAK2 and STAT3 were observed. CONCLUSION These results indicated that ELP reduced the severity of arthritis and joint swelling, suggesting an antirheumatic effect on CIA rats. The possible mechanism is related to inhibiting inflammatory response and inducing apoptosis in synovial tissues by regulating JAK2/STAT3 signaling pathway. However, further in vivo and in vitro investigations are still needed to clarify the underlying mechanism of ELP in treating RA.
Collapse
Affiliation(s)
- Chuan Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qian Zhao
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lu Zhong
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiuyue Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Rui Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shuang Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yangxin Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ning Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jinsong Su
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wüntrang Dhondrup
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ya Tu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Development Research Center of Traditional Chinese Medicine, China Academy of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
21
|
Gallic acid: Pharmacological activities and molecular mechanisms involved in inflammation-related diseases. Biomed Pharmacother 2021; 133:110985. [DOI: 10.1016/j.biopha.2020.110985] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
|
22
|
Aghakhani S, Zerrouk N, Niarakis A. Metabolic Reprogramming of Fibroblasts as Therapeutic Target in Rheumatoid Arthritis and Cancer: Deciphering Key Mechanisms Using Computational Systems Biology Approaches. Cancers (Basel) 2020; 13:E35. [PMID: 33374292 PMCID: PMC7795338 DOI: 10.3390/cancers13010035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/29/2022] Open
Abstract
Fibroblasts, the most abundant cells in the connective tissue, are key modulators of the extracellular matrix (ECM) composition. These spindle-shaped cells are capable of synthesizing various extracellular matrix proteins and collagen. They also provide the structural framework (stroma) for tissues and play a pivotal role in the wound healing process. While they are maintainers of the ECM turnover and regulate several physiological processes, they can also undergo transformations responding to certain stimuli and display aggressive phenotypes that contribute to disease pathophysiology. In this review, we focus on the metabolic pathways of glucose and highlight metabolic reprogramming as a critical event that contributes to the transition of fibroblasts from quiescent to activated and aggressive cells. We also cover the emerging evidence that allows us to draw parallels between fibroblasts in autoimmune disorders and more specifically in rheumatoid arthritis and cancer. We link the metabolic changes of fibroblasts to the toxic environment created by the disease condition and discuss how targeting of metabolic reprogramming could be employed in the treatment of such diseases. Lastly, we discuss Systems Biology approaches, and more specifically, computational modeling, as a means to elucidate pathogenetic mechanisms and accelerate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Sahar Aghakhani
- GenHotel, University of Evry, University of Paris-Saclay, Genopole, 91000 Evry, France; (S.A.); (N.Z.)
- Lifeware Group, Inria Saclay, 91120 Palaiseau, France
| | - Naouel Zerrouk
- GenHotel, University of Evry, University of Paris-Saclay, Genopole, 91000 Evry, France; (S.A.); (N.Z.)
| | - Anna Niarakis
- GenHotel, University of Evry, University of Paris-Saclay, Genopole, 91000 Evry, France; (S.A.); (N.Z.)
- Lifeware Group, Inria Saclay, 91120 Palaiseau, France
| |
Collapse
|
23
|
Masoumi M, Mehrabzadeh M, Mahmoudzehi S, Mousavi MJ, Jamalzehi S, Sahebkar A, Karami J. Role of glucose metabolism in aggressive phenotype of fibroblast-like synoviocytes: Latest evidence and therapeutic approaches in rheumatoid arthritis. Int Immunopharmacol 2020; 89:107064. [PMID: 33039953 DOI: 10.1016/j.intimp.2020.107064] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/10/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Glucose metabolism is considerably increased in inflamed joints of rheumatoid arthritis (RA) patients at early stages. Fibroblast-like synoviocytes (FLSs) activation and subsequent joint damage are linked with metabolic alterations, especially glucose metabolism. It has been shown that glucose metabolism is elevated in aggressive phenotype of FLS cells. In this regard, glycolytic blockers are able to reduce aggressiveness of the FLS cells resulting in decreased joint damage in various arthritis models. Besides, metabolic changes in immune and non-immune cells such as FLS can provide important targets for therapeutic intervention. Glycolytic enzymes such as hexokinase 2 (HK2), phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB), and phosphoglycerate kinase (PGK) play essential roles in aggressive behavior of FLS cells. It has been documented that the HK2 enzyme is significantly upregulated in RA FLS cells, compared with osteoarthritis (OA) FLS cells. The HK2 is expressed in a few tissues and upregulated in the inflamed synovium of RA patients that makes it a potential target for RA treatment. Furthermore, HK2 has different roles in each cellular compartment, which offers another level of specificity and provides a specific target to reduce deleterious effects of inhibiting the enzyme in RA without affecting glycolysis in normal cells. Thus, targeting the HK2 enzyme might be an attractive potential selective target for arthritis therapy and safer than global glycolysis inhibition. Therefore, this review was aimed to summarize the current knowledge about glucose metabolism of FLS cells and suggest novel biomarkers, which are potential candidates for RA treatment.
Collapse
Affiliation(s)
- Maryam Masoumi
- Clinical Research Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran
| | - Mohsen Mehrabzadeh
- Department of Medical Biochemistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Salman Mahmoudzehi
- Department of Medical Laboratory Sciences, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sirous Jamalzehi
- Department of Medical Laboratory Sciences, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Halal Research Center of IRI, FDA, Tehran, Iran.
| | - Jafar Karami
- Department of Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Wu W, Zhang Z, Chao Z, Li B, Li R, Jiang A, Kim KH, Liu H. Transcriptome analysis reveals the genetic basis of skeletal muscle glycolytic potential based on a pig model. Gene 2020; 766:145157. [PMID: 32949697 DOI: 10.1016/j.gene.2020.145157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/13/2020] [Accepted: 09/11/2020] [Indexed: 10/23/2022]
Abstract
Glycolytic potential (GP) calculated based on glucose, glycogen, glucose-6-phosphate, and lactate contents is a critical factor for multiple meat quality characteristics. However, the genetic basis of glycolytic metabolism is still unclear. In this study, we constructed six RNA-Seq libraries using longissimus dorsi (LD) muscles from pigs divergent for GP phenotypic values and generated the whole genome-wide gene expression profiles. Furthermore, we identified 25,880 known and 220 novel genes from these skeletal muscle libraries, and 222 differentially expressed genes (DEGs) between the higher and lower GP groups. Notably, we found that the Lactate dehydrogenase B (LDHB) and Fructose-2, 6-biphosphatase 3 (PFKFB3) expression levels were higher in the higher GP group than the lower GP group, and positively correlated with GP and lactic acid (LA), and reversely correlated with pH value at 45 min postmortem (pH45min). Besides, LDHB and PFKFB3 expression were positively correlated with drip loss measured at 48 h postmortem (DL48h) and drip loss measured at 24 h postmortem (DL24h). Collectively, we identified a serial of DEGs as the potential key candidate genes affecting GP and found that LDHB and PFKFB3 are closely related to GP and GP-related traits. Our results lay a solid basis for in-depth studies of the regulatory mechanisms on GP and GP-related traits in pigs.
Collapse
Affiliation(s)
- Wangjun Wu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| | - Zengkai Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhe Chao
- Institute of Animal Science & Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Bojiang Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Rongyang Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Aiwen Jiang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kee-Hong Kim
- Department of Food Science, Purdue University, West Lafayette, IN 47897, USA.
| | - Honglin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
25
|
Du H, Wang Y, Zeng Y, Huang X, Liu D, Ye L, Li Y, Chen X, Liu T, Li H, Wu J, Yu Q, Wu Y, Jie L. Tanshinone IIA Suppresses Proliferation and Inflammatory Cytokine Production of Synovial Fibroblasts from Rheumatoid Arthritis Patients Induced by TNF-α and Attenuates the Inflammatory Response in AIA Mice. Front Pharmacol 2020; 11:568. [PMID: 32499694 PMCID: PMC7243269 DOI: 10.3389/fphar.2020.00568] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/14/2020] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive autoimmune disease in which activated RA fibroblast-1ike synoviocytes (RA-FLSs) are one of the main factors responsible for inducing morbidity. Previous reports have shown that RA-FLSs have proliferative features similar to cancer cells, in addition to causing cartilage erosion that eventually causes joint damage. Thus, new therapeutic strategies and drugs that can effectively contain the abnormal hyperplasia of RA-FLSs and restrain RA development are necessary for the treatment of RA. Tanshinone IIA (Tan IIA), one of the main phytochemicals isolated from Salvia miltiorrhiza Bunge, is capable of promoting RA-FLS apoptosis and inhibiting arthritis in an AIA mouse model. In addition, RA patients treated at our clinic with Tan IIA showed significant improvements in their clinical symptoms. However, the details of the molecular mechanism by which Tan IIA effects RA are unknown. To clarify this mechanism, we evaluated the antiproliferative and inhibitory effects of proinflammatory factor production caused by Tan IIA to RA-FLSs. We demonstrated that Tan IIA can restrict the proliferation, migration, and invasion of RA-FLSs in a time- and dose-dependent manner. Moreover, Tan IIA effectively suppressed the increase in mRNA expression of some matrix metalloproteinases and proinflammatory factors induced by TNF-α in RA-FLSs, resulting in inflammatory reactivity inhibition and blocking the destruction of the knee joint. Through the integration of network pharmacology analyses with the experimental data obtained, it is revealed that the effects of Tan IIA on RA can be attributed to its influence on different signaling pathways, including MAPK, AKT/mTOR, HIF-1, and NF-kB. Taken together, these data suggest that the compound Tan IIA has great therapeutic potential for RA treatment.
Collapse
Affiliation(s)
- Hongyan Du
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yuechun Wang
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yongchang Zeng
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoming Huang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Dingfei Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Lvlan Ye
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yang Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaochen Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Hongwei Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jing Wu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qinghong Yu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingsong Wu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Ligang Jie
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Tseng CC, Wu LY, Tsai WC, Ou TT, Wu CC, Sung WY, Kuo PL, Yen JH. Differential Expression Profiles of the Transcriptome and miRNA Interactome in Synovial Fibroblasts of Rheumatoid Arthritis Revealed by Next Generation Sequencing. Diagnostics (Basel) 2019; 9:diagnostics9030098. [PMID: 31426562 PMCID: PMC6787660 DOI: 10.3390/diagnostics9030098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Using next-generation sequencing to decipher the molecular mechanisms underlying aberrant rheumatoid arthritis synovial fibroblasts (RASF) activation, we performed transcriptome-wide RNA-seq and small RNA-seq on synovial fibroblasts from rheumatoid arthritis (RA) subject and normal donor. Differential expression of mRNA and miRNA was integrated with interaction analysis, functional annotation, regulatory network mapping and experimentally verified miRNA–target interaction data, further validated with microarray expression profiles. In this study, 3049 upregulated mRNA and 3552 downregulated mRNA, together with 50 upregulated miRNA and 35 downregulated miRNA in RASF were identified. Interaction analysis highlighted contribution of miRNA to altered transcriptome. Functional annotation revealed metabolic deregulation and oncogenic signatures of RASF. Regulatory network mapping identified downregulated FOXO1 as master transcription factor resulting in altered transcriptome of RASF. Differential expression in three miRNA and corresponding targets (hsa-miR-31-5p:WASF3, hsa-miR-132-3p:RB1, hsa-miR-29c-3p:COL1A1) were also validated. The interactions of these three miRNA–target genes were experimentally validated with past literature. Our transcriptomic and miRNA interactomic investigation identified gene signatures associated with RASF and revealed the involvement of transcription factors and miRNA in an altered transcriptome. These findings help facilitate our understanding of RA with the hope of serving as a springboard for further discoveries relating to the disease.
Collapse
Affiliation(s)
- Chia-Chun Tseng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Ling-Yu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wen-Chan Tsai
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Tsan-Teng Ou
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Cheng-Chin Wu
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Wan-Yu Sung
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan.
| | - Jeng-Hsien Yen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|
27
|
de Oliveira PG, Farinon M, Sanchez-Lopez E, Miyamoto S, Guma M. Fibroblast-Like Synoviocytes Glucose Metabolism as a Therapeutic Target in Rheumatoid Arthritis. Front Immunol 2019; 10:1743. [PMID: 31428089 PMCID: PMC6688519 DOI: 10.3389/fimmu.2019.01743] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/10/2019] [Indexed: 12/29/2022] Open
Abstract
Metabolomic studies show that rheumatoid arthritis (RA) is associated with metabolic disruption that may be therapeutically targetable. Among them, glucose metabolism and glycolytic intermediaries seem to have an important role in fibroblast-like synoviocytes (FLS) phenotype and might contribute to early stage disease pathogenesis. RA FLS are transformed from quiescent to aggressive and metabolically active cells and several works have shown that glucose metabolism is increased in activated FLS. Glycolytic inhibitors reduce not only FLS aggressive phenotype in vitro but also decrease bone and cartilage damage in several murine models of arthritis. Essential glycolytic enzymes, including hexokinase 2 (HK2) and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase (PFKFB) enzymes, have important roles in FLS behavior. Of interest, HK2 is an inducible enzyme present only in the inflamed rheumatic tissues compared to osteoarthritis synovium. It is a contributor to glucose metabolism that could be selectively targeted without compromising systemic homeostasis as a novel approach for combination therapy independent of systemic immunosuppression. More information about metabolic targets that do not compromise global glucose metabolism in normal cells is needed.
Collapse
Affiliation(s)
| | - Mirian Farinon
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Elsa Sanchez-Lopez
- Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Shigeki Miyamoto
- Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Monica Guma
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
28
|
Du H, Zhang X, Zeng Y, Huang X, Chen H, Wang S, Wu J, Li Q, Zhu W, Li H, Liu T, Yu Q, Wu Y, Jie L. A Novel Phytochemical, DIM, Inhibits Proliferation, Migration, Invasion and TNF-α Induced Inflammatory Cytokine Production of Synovial Fibroblasts From Rheumatoid Arthritis Patients by Targeting MAPK and AKT/mTOR Signal Pathway. Front Immunol 2019; 10:1620. [PMID: 31396207 PMCID: PMC6663984 DOI: 10.3389/fimmu.2019.01620] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/28/2019] [Indexed: 01/19/2023] Open
Abstract
In rheumatoid arthritis(RA) pathogenesis, activated RA fibroblast-like synoviocytes (RA-FLSs) exhibit similar proliferative features as tumor cells and subsequent erosion to cartilage will eventually lead to joint destruction. Therefore, it is imperative to search for compounds, which can effectively inhibit the abnormal activation of RA-FLSs, and retard RA progression.3′3-Diindolylmethane (DIM), the major product of the acid-catalyzed oligomerization of indole-3-carbinol from cruciferous vegetables, has been reported to be functionally relevant to inhibition of migration, invasion and carcinogenesis in some solid tumors. In this study, we explored the anti-proliferation, anti-metastasis and anti-inflammation effects of DIM on RA-FLSs as well as the underlying molecular mechanisms. To do this, primary RA-FLSs were isolated from RA patients and an animal model. Cell proliferation, migration and invasion were measured using CCK-8, scratch, and Transwell assays, respectively. The effects of DIM on Matrix metalloproteinases (MMPs) and some inflammatory factors mRNA and key molecules such as some inflammatory factors and those involved in aberrantly-activated signaling pathway in response to tumor necrosis factor α(TNF-α), a typical characteristic mediator in RA-FLS, were quantitatively measured by real-time PCR and western blotting. Moreover, the effect of DIM on adjuvant induced arthritis(AIA) models was evaluated with C57BL/6 mice in vivo. The results showed that DIM inhibited proliferation, migration and invasion of RA-FLS in vitro. Meanwhile, DIM dramatically suppressed TNF-α–induced increases in the mRNA levels of MMP-2, MMP-3, MMP-8, and MMP-9; as well as the proinflammatory factors IL-6, IL-8, and IL-1β. Mechanistic studies revealed that DIM is able to suppress phosphorylated activation not only of p38, JNK in MAPK pathway but of AKT, mTOR and downstream molecules in the AKT/mTOR pathway. Moreover, DIM treatment decreased expression levels of proinflammatory cytokines in the serum and alleviated arthritis severity in the knee joints of AIA mice. Taken together, our findings demonstrate that DIM could inhibit proliferation, migration and invasion of RA-FLSs and reduce proinflammatory factors induced by TNF-α in vitro by blocking MAPK and AKT/mTOR pathway and prevent inflammation and knee joint destruction in vivo, which suggests that DIM might have therapeutic potential for RA.
Collapse
Affiliation(s)
- Hongyan Du
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xi Zhang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yongchang Zeng
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoming Huang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Hao Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Suihai Wang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jing Wu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Li
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Zhu
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Hongwei Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qinghong Yu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingsong Wu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Ligang Jie
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|