1
|
Liu X, Zheng Y, Li H, Ma Y, Cao R, Zheng Z, Tian Y, Du L, Zhang J, Zhang C, Gao J. The role of metabolites in the progression of osteoarthritis: Mechanisms and advances in therapy. J Orthop Translat 2025; 50:56-70. [PMID: 39868350 PMCID: PMC11762942 DOI: 10.1016/j.jot.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 01/28/2025] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disease affected by many factors, and there is currently no effective treatment. In recent years, the latest progress in metabolomics in OA research has revealed several metabolic pathways and new specific metabolites involved in OA. Metabolites play significant roles in the identification and management of OA. This review looks back on the development history of metabolomics and the progress of this technology in OA as well as its potential clinical applications. It summarizes the applications of metabolites in the field of OA and future research directions. This understanding will advance the identification of metabolic treatment goals for OA. The translational potential of this article The development of metabolomics offers possibilities for the treatment of OA. This article reviews the relationship between metabolites associated with chondrocytes and OA. Selectively altering these three metabolic pathways and their associated metabolites may hold great potential as new focal points for OA treatment.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Yongqiang Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ruomu Cao
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuchen Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College
| | - Jinshan Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| |
Collapse
|
2
|
Hu S, Lin Y, Tang Y, Zhang J, He Y, Li G, Li L, Cai X. Targeting dysregulated intracellular immunometabolism within synovial microenvironment in rheumatoid arthritis with natural products. Front Pharmacol 2024; 15:1403823. [PMID: 39104392 PMCID: PMC11298361 DOI: 10.3389/fphar.2024.1403823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Immunometabolism has been an emerging hotspot in the fields of tumors, obesity, and atherosclerosis in recent decades, yet few studies have investigated its connection with rheumatoid arthritis (RA). In principle, intracellular metabolic pathways upstream regulated by nutrients and growth factors control the effector functions of immune cells. Dynamic communication and hypermetabolic lesions of immune cells within the inflammatory synovial microenvironment contributes to the development and progression of RA. Hence, targeting metabolic pathways within immune subpopulations and pathological cells may represent novel therapeutic strategies for RA. Natural products constitute a great potential treasury for the research and development of novel drugs targeting RA. Here, we aimed to delineate an atlas of glycolysis, lipid metabolism, amino acid biosynthesis, and nucleotide metabolism in the synovial microenvironment of RA that affect the pathological processes of synovial cells. Meanwhile, therapeutic potentials and pharmacological mechanisms of natural products that are demonstrated to inhibit related key enzymes in the metabolic pathways or reverse the metabolic microenvironment and communication signals were discussed and highlighted.
Collapse
Affiliation(s)
- Shengtao Hu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ye Lin
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuanyuan Tang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junlan Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yini He
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gejing Li
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liqing Li
- The Central Research Laboratory, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Central Research Laboratory, Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China
| |
Collapse
|
3
|
Adam MS, Zhuang H, Ren X, Zhang Y, Zhou P. The metabolic characteristics and changes of chondrocytes in vivo and in vitro in osteoarthritis. Front Endocrinol (Lausanne) 2024; 15:1393550. [PMID: 38854686 PMCID: PMC11162117 DOI: 10.3389/fendo.2024.1393550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
Osteoarthritis (OA) is an intricate pathological condition that primarily affects the entire synovial joint, especially the hip, hand, and knee joints. This results in inflammation in the synovium and osteochondral injuries, ultimately causing functional limitations and joint dysfunction. The key mechanism responsible for maintaining articular cartilage function is chondrocyte metabolism, which involves energy generation through glycolysis, oxidative phosphorylation, and other metabolic pathways. Some studies have shown that chondrocytes in OA exhibit increased glycolytic activity, leading to elevated lactate production and decreased cartilage matrix synthesis. In OA cartilage, chondrocytes display alterations in mitochondrial activity, such as decreased ATP generation and increased oxidative stress, which can contribute to cartilage deterioration. Chondrocyte metabolism also involves anabolic processes for extracellular matrix substrate production and energy generation. During OA, chondrocytes undergo considerable metabolic changes in different aspects, leading to articular cartilage homeostasis deterioration. Numerous studies have been carried out to provide tangible therapies for OA by using various models in vivo and in vitro targeting chondrocyte metabolism, although there are still certain limitations. With growing evidence indicating the essential role of chondrocyte metabolism in disease etiology, this literature review explores the metabolic characteristics and changes of chondrocytes in the presence of OA, both in vivo and in vitro. To provide insight into the complex metabolic reprogramming crucial in chondrocytes during OA progression, we investigate the dynamic interaction between metabolic pathways, such as glycolysis, lipid metabolism, and mitochondrial function. In addition, this review highlights prospective future research directions for novel approaches to diagnosis and treatment. Adopting a multifaceted strategy, our review aims to offer a comprehensive understanding of the metabolic intricacies within chondrocytes in OA, with the ultimate goal of identifying therapeutic targets capable of modulating chondrocyte metabolism for the treatment of OA.
Collapse
Affiliation(s)
| | | | | | | | - Panghu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Li H, Cui Y, Wang J, Zhang W, Chen Y, Zhao J. Identification and validation of biomarkers related to lipid metabolism in osteoarthritis based on machine learning algorithms. Lipids Health Dis 2024; 23:111. [PMID: 38637751 PMCID: PMC11025229 DOI: 10.1186/s12944-024-02073-5] [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: 01/29/2024] [Accepted: 03/07/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Osteoarthritis and lipid metabolism are strongly associated, although the precise targets and regulatory mechanisms are unknown. METHODS Osteoarthritis gene expression profiles were acquired from the GEO database, while lipid metabolism-related genes (LMRGs) were sourced from the MigSB database. An intersection was conducted between these datasets to extract gene expression for subsequent differential analysis. Following this, functional analyses were performed on the differentially expressed genes (DEGs). Subsequently, machine learning was applied to identify hub genes associated with lipid metabolism in osteoarthritis. Immune-infiltration analysis was performed using CIBERSORT, and external datasets were employed to validate the expression of these hub genes. RESULTS Nine DEGs associated with lipid metabolism in osteoarthritis were identified. UGCG and ESYT1, which are hub genes involved in lipid metabolism in osteoarthritis, were identified through the utilization of three machine learning algorithms. Analysis of the validation dataset revealed downregulation of UGCG in the experimental group compared to the normal group and upregulation of ESYT1 in the experimental group compared to the normal group. CONCLUSIONS UGCG and ESYT1 were considered as hub LMRGs in the development of osteoarthritis, which were regarded as candidate diagnostic markers. The effects are worth expected in the early diagnosis and treatment of osteoarthritis.
Collapse
Affiliation(s)
- Hang Li
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Yubao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Jian Wang
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Wei Zhang
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Yuhao Chen
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Jijun Zhao
- Department of Orthopedic, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China.
| |
Collapse
|
5
|
Wang Y, Xie H, Huang X, Chen K, Zhu Y, Yao G. Retrospective analysis and preliminary laboratory validation of treatment efficacy and blood lipid levels in patients with rheumatoid arthritis. Clin Rheumatol 2023; 42:3213-3223. [PMID: 37488372 DOI: 10.1007/s10067-023-06683-9] [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: 01/03/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
OBJECTIVE Rheumatoid arthritis is a systemic disease characterized by progressive chronic inflammation resulting in destruction of synovial joints. In addition to joint involvement, abnormal blood lipid indexes have also been found in RA patients. The correlation between various blood lipid indexes and the treatment effects were assessed in patients with rheumatoid arthritis, for the purposes to find a better medication strategy for RA. METHODS One hundred nineteen rheumatoid arthritis patients were recruited and divided into two groups, 45 patients with significant drug treatment effect and 45 patients with insignificant drug treatment effect through the nearest neighbor matching method in propensity score. The correlation between various blood lipid indexes and drug treatment effect of rheumatoid arthritis patients was analyzed. A mouse model of rheumatoid arthritis was constructed in the laboratory; methotrexate was treated as a positive drug. We observe and record the onset of rheumatoid arthritis in mice, as well as the proportion of immune cells, the expression of inflammatory factors, and the changes in blood lipid profiles was done. RESULTS The levels of total cholesterol (TC), triglyceride (TG), high density lipoprotein (HDL-C), low density lipoprotein (LDL-C), and erythrocyte sedimentation rate (ESR) in rheumatoid arthritis patients were significantly different between the two groups (P < 0.05). There was no significant difference in other indexes between the two groups (P > 0.05). Methotrexate had a good therapeutic effect on CIA model mice, and the levels of TC and HDL-C in the treatment group were higher than those in the model group. CONCLUSION There is a high correlation between the levels of TC and HDL-C in rheumatoid arthritis patients and the effect of drug treatment. In the clinical treatment of rheumatoid arthritis, we should focus on improving the blood lipid indexes such as TC and HDL-C, and explore more targeted individualized administration, so as to achieve better and faster treatment effect in patients with rheumatoid arthritis. Key Points • In this research, we found that the TC and HDL-C level in RA patients' blood is highly related with the therapeutic effect, and a lower level of TC and HDL-C is better for therapeutic effect of RA.
Collapse
Affiliation(s)
- Yuchun Wang
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| | - Han Xie
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| | - Xuan Huang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| | - Keyuan Chen
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China.
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| |
Collapse
|
6
|
Wang Z, Wang M, Xu X, Liu Y, Chen Q, Wu B, Zhang Y. PPARs/macrophages: A bridge between the inflammatory response and lipid metabolism in autoimmune diseases. Biochem Biophys Res Commun 2023; 684:149128. [PMID: 39491979 DOI: 10.1016/j.bbrc.2023.149128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 11/05/2024]
Abstract
Autoimmune diseases (AIDs) are a collection of pathologies that arise from autoimmune reactions and lead to the destruction and damage of the body's tissues and cellular components, ultimately resulting in tissue damage and organ dysfunction. The anti-inflammatory effects of the peroxisome proliferator-activated receptor (PPAR), a pivotal regulator of lipid metabolism, are crucial in the context of AIDs. PPAR mitigates AIDs by modulating macrophage polarization and suppressing the inflammatory response. Numerous studies have demonstrated the crucial involvement of lipid metabolism and phenotypic switching in classically activated (M1)/alternatively activated (M2)-like macrophages in the inflammatory pathway of AIDs. However, the precise mechanism by which PPAR, a critical mediator between of lipid metabolism and macrophage polarization, regulates macrophage polarization remains unclear. This review aimed to clarify the role of PPAR and macrophages in the triangular relationship among AIDs, lipid metabolism, and inflammatory response, and aims to summarize the mechanism of the PPAR-mediated macrophage activation and polarization, which impacts the progression and development of AIDs.
Collapse
Affiliation(s)
- Zikang Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401334, China
| | - Miao Wang
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Xiaoyu Xu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401334, China; Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Yunyan Liu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401334, China
| | - Qian Chen
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401334, China
| | - Bin Wu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401334, China; Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China.
| | - Ying Zhang
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 401334, China; Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China.
| |
Collapse
|
7
|
Lei Q, Yang J, Li L, Zhao N, Lu C, Lu A, He X. Lipid metabolism and rheumatoid arthritis. Front Immunol 2023; 14:1190607. [PMID: 37325667 PMCID: PMC10264672 DOI: 10.3389/fimmu.2023.1190607] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
As a chronic progressive autoimmune disease, rheumatoid arthritis (RA) is characterized by mainly damaging the synovium of peripheral joints and causing joint destruction and early disability. RA is also associated with a high incidence rate and mortality of cardiovascular disease. Recently, the relationship between lipid metabolism and RA has gradually attracted attention. Plasma lipid changes in RA patients are often detected in clinical tests, the systemic inflammatory status and drug treatment of RA patients can interact with the metabolic level of the body. With the development of lipid metabolomics, the changes of lipid small molecules and potential metabolic pathways have been gradually discovered, which makes the lipid metabolism of RA patients or the systemic changes of lipid metabolism after treatment more and more comprehensive. This article reviews the lipid level of RA patients, as well as the relationship between inflammation, joint destruction, cardiovascular disease, and lipid level. In addition, this review describes the effect of anti-rheumatic drugs or dietary intervention on the lipid profile of RA patients to better understand RA.
Collapse
Affiliation(s)
- Qian Lei
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Yang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Gong Y, Li S, Wu J, Zhang T, Fang S, Feng D, Luo X, Yuan J, Wu Y, Yan X, Zhang Y, Zhu J, Wu J, Lian J, Xiang W, Ni Z. Autophagy in the pathogenesis and therapeutic potential of post-traumatic osteoarthritis. BURNS & TRAUMA 2023; 11:tkac060. [PMID: 36733467 PMCID: PMC9887948 DOI: 10.1093/burnst/tkac060] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Indexed: 02/04/2023]
Abstract
Autophagy, as a fundamental mechanism for cellular homeostasis, is generally involved in the occurrence and progression of various diseases. Osteoarthritis (OA) is the most common musculoskeletal disease that often leads to pain, disability and economic loss in patients. Post-traumatic OA (PTOA) is a subtype of OA, accounting for >12% of the overall burden of OA. PTOA is often caused by joint injuries including anterior cruciate ligament rupture, meniscus tear and intra-articular fracture. Although a variety of methods have been developed to treat acute joint injury, the current measures have limited success in effectively reducing the incidence and delaying the progression of PTOA. Therefore, the pathogenesis and intervention strategy of PTOA need further study. In the past decade, the roles and mechanisms of autophagy in PTOA have aroused great interest in the field. It was revealed that autophagy could maintain the homeostasis of chondrocytes, reduce joint inflammatory level, prevent chondrocyte death and matrix degradation, which accordingly improved joint symptoms and delayed the progression of PTOA. Moreover, many strategies that target PTOA have been revealed to promote autophagy. In this review, we summarize the roles and mechanisms of autophagy in PTOA and the current strategies for PTOA treatment that depend on autophagy regulation, which may be beneficial for PTOA patients in the future.
Collapse
Affiliation(s)
| | | | | | - Tongyi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China,Department of General practice, Chinese PLA General Hospital of the Central Theater Command, Wuluo Street, Wuchang District, Wuhan 430000, China
| | - Shunzheng Fang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China
| | - Daibo Feng
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China
| | - Xiaoqing Luo
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Street, Yuzhong District, Chongqing 400042, China
| | - Jing Yuan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing 400038, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing 400038, China
| | - Xiaojing Yan
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing 400038, China
| | - Yan Zhang
- Department of Pediatrics, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Guoben Street, Wanzhou district, Chongqing 404000, China
| | - Jun Zhu
- Department of Cardiology, Shanghai Hospital, Shanghai Street, Wanzhou District, Chongqing 404000, China
| | - Jiangyi Wu
- Department of Sports Medicine and Rehabilitation, Shenzhen Hospital, Peking University, Lianhua Street, Futian District, Shenzhen 518034, China
| | - Jiqin Lian
- Correspondence. Zhenghong Ni, ; Wei Xiang, ; Jiqin Lian,
| | - Wei Xiang
- Correspondence. Zhenghong Ni, ; Wei Xiang, ; Jiqin Lian,
| | - Zhenhong Ni
- Correspondence. Zhenghong Ni, ; Wei Xiang, ; Jiqin Lian,
| |
Collapse
|
9
|
Chen Y, Zhu XW, Lai WF, Liu YP, Xu XF, Liu LM, Chen YJ, Zhang CF, Wang GY, Cheng ZQ, Liu DZ. Gancao Nourishing-Yin decoction combined with methotrexate in treatment of aging CIA mice: a study based on DIA proteomic analysis. Chin Med 2023; 18:9. [PMID: 36709303 PMCID: PMC9883815 DOI: 10.1186/s13020-023-00709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/04/2023] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Elderly rheumatoid arthritis (ERA) population faces multiple treatment dilemma. Here we aim to investigate if Gancao Nourishing-Yin decoction (GCNY) added to methotrexate (MTX) exhibit better effects in an ERA mice model. METHODS ERA mice model was established by adding D-galactose (Dgal) to collagen-induced arthritis (CIA) mice. The model was then assigned into control group (CIA + Dgal), MTX treatment group (MTX), GCNY treatment group (GCNY), and integrative treatment group (MTX + GCNY). Pathological scoring was performed to evaluate the severity between the groups. Proteomic analysis was applied to investigate the secretory phenotype of the ERA mouse model and the underlying mechanism of GCNY, MTX and their combination. Representative cytokines related to proteomic results were further validated by ELISAs. RESULTS CIA + Dgal mice showed more aggressive joints damage than the CIA mice. Besides changes in the inflammatory pathway such as Pi3k-Akt signaling pathway in both model, differential expressed proteins (DEPs) indicated metabolism-related pathways were more obvious in CIA + Dgal mice. Low-dose MTX failed to show pathological improvement in CIA + Dgal mice, while GCNY improved joints damage significantly. Besides down-regulated inflammation-related targets, GCNY-regulated DEPs (such as Apoc1 ~ 3, Grk2 and Creb3l3) were broadly enriched in metabolism-related pathways. MTX + GCNY showed the best therapeutic effect, and the DEPs enriched in a variety of inflammatory,metabolism and osteoclast differentiation signaling pathway. Notably, MTX + GCNY treatment up-regulated Dhfr, Cbr1, Shmt1 involved in folic acid biosynthesis and anti-folate resistance pathways indicated a coincidence synergic action. ELISAs confirmed CPR and Akt that elevated in CIA + Dgal mice were significantly ameliorated by treatments, and adding on GCNY elevated folic acid levels and its regulator Dhfr. CONCLUSION Aging aggravated joints damage in CIA, which probably due to metabolic changes rather than more severe inflammation. GCNY showed significant effects in the ERA mice model especially when integrated with MTX to obtain a synergic action.
Collapse
Affiliation(s)
- Yong Chen
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China ,grid.440218.b0000 0004 1759 7210Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People‘s Hospital, Shenzhen, China
| | - Xing-wang Zhu
- grid.284723.80000 0000 8877 7471Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Wing-Fu Lai
- grid.16890.360000 0004 1764 6123Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong, Hong Kong Special Administrative Region China ,Department of Urology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang China
| | - Yong-pu Liu
- grid.284723.80000 0000 8877 7471Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Xuan-feng Xu
- grid.284723.80000 0000 8877 7471Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Li-ming Liu
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| | - Yan-juan Chen
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| | - Chuan-fu Zhang
- grid.412540.60000 0001 2372 7462The Seventh People’s Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-yi Wang
- grid.413458.f0000 0000 9330 9891Guizhou Medical University affiliated Hospital, Guiyang, China
| | - Zhi-qiang Cheng
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| | - Dong-zhou Liu
- grid.440218.b0000 0004 1759 7210Shenzhen People‘s Hospital (The Second Clinical Medical College of Jinan University and the First Affiliated Hospital to Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
10
|
Chen X, Liu J, Sun Y, Wen J, Zhou Q, Ding X, Zhang X. Correlation analysis of differentially expressed long non-coding RNA HOTAIR with PTEN/PI3K/AKT pathway and inflammation in patients with osteoarthritis and the effect of baicalin intervention. J Orthop Surg Res 2023; 18:34. [PMID: 36635778 PMCID: PMC9835332 DOI: 10.1186/s13018-023-03505-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE This study aims to investigate the correlation of long non-coding RNA HOX transcript antisense RNA (lncRNA HOTAIR) with the PTEN/PI3K/AKT pathway and clinical-related indicators in osteoarthritis (OA) and determine the effect of baicalin intervention. METHODS The levels of clinical lipid metabolism indexes and immune-inflammatory indexes in OA patients and normal controls was detected. OA chondrocytes (OA-CHs) were induced with peripheral blood mononuclear cells (PBMCs), followed by baicalin treatment (50 ug/mL). RT-qPCR was performed to measure lncRNA HOTAIR expression. The levels of inflammatory cytokines and adiponectin were detected using ELISA kits. CCK-8 assay was used to assess the viability of CHs. The related protein expression was measured using Western blot analysis. RESULTS LncRNA HOTAIR might act as a biomarker of OA in vivo. LncRNA HOTAIR was positively correlated with TC, hs-CRP, IgA, TNF-α, and VAS score. Overexpression of lncRNA HOTAIR in vitro inhibited cell proliferation, reduced IL-10 and PTEN expression, but augmented TNF-α, p-PI3K, and p-AKT proteins in OA-CHs stimulated by OA-PBMCs. The changes of above indexes were also observed in OA-CHs stimulated by OA-PBMCs treated with si-lncRNA HOTAIR or baicalin, implying the synergistic effects of baicalin and lncRNA HOTAIR silencing on OA. CONCLUSIONS Conclusively, lncRNA HOTAIR was highly expressed in OA-CHs, which facilitated OA inflammatory responses by orchestrating inflammatory cytokines and the PTEN/PI3K/AKT pathway. Baicalin exerted therapeutic effects by inhibiting the expression of lncRNA HOTAIR, decreasing the protein levels of p-PI3K and p-AKT, and increasing the protein levels of PTEN, APN, and ADIPOR1.
Collapse
Affiliation(s)
- Xiaolu Chen
- grid.252251.30000 0004 1757 8247Anhui University of Traditional Chinese Medicine, Hefei, 230031 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| | - Jian Liu
- grid.412679.f0000 0004 1771 3402Department of Rheumatology and Immunology, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230038 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| | - Yanqiu Sun
- grid.252251.30000 0004 1757 8247Anhui University of Traditional Chinese Medicine, Hefei, 230031 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| | - Jianting Wen
- grid.252251.30000 0004 1757 8247Anhui University of Traditional Chinese Medicine, Hefei, 230031 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| | - Qin Zhou
- grid.252251.30000 0004 1757 8247Anhui University of Traditional Chinese Medicine, Hefei, 230031 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| | - Xiang Ding
- grid.252251.30000 0004 1757 8247Anhui University of Traditional Chinese Medicine, Hefei, 230031 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| | - Xianheng Zhang
- grid.252251.30000 0004 1757 8247Anhui University of Traditional Chinese Medicine, Hefei, 230031 Anhui Province China ,grid.252251.30000 0004 1757 8247Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, 230012 Anhui Province China
| |
Collapse
|
11
|
Gurke R, Bendes A, Bowes J, Koehm M, Twyman RM, Barton A, Elewaut D, Goodyear C, Hahnefeld L, Hillenbrand R, Hunter E, Ibberson M, Ioannidis V, Kugler S, Lories RJ, Resch E, Rüping S, Scholich K, Schwenk JM, Waddington JC, Whitfield P, Geisslinger G, FitzGerald O, Behrens F, Pennington SR. Omics and Multi-Omics Analysis for the Early Identification and Improved Outcome of Patients with Psoriatic Arthritis. Biomedicines 2022; 10:2387. [PMID: 36289648 PMCID: PMC9598654 DOI: 10.3390/biomedicines10102387] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
The definitive diagnosis and early treatment of many immune-mediated inflammatory diseases (IMIDs) is hindered by variable and overlapping clinical manifestations. Psoriatic arthritis (PsA), which develops in ~30% of people with psoriasis, is a key example. This mixed-pattern IMID is apparent in entheseal and synovial musculoskeletal structures, but a definitive diagnosis often can only be made by clinical experts or when an extensive progressive disease state is apparent. As with other IMIDs, the detection of multimodal molecular biomarkers offers some hope for the early diagnosis of PsA and the initiation of effective management and treatment strategies. However, specific biomarkers are not yet available for PsA. The assessment of new markers by genomic and epigenomic profiling, or the analysis of blood and synovial fluid/tissue samples using proteomics, metabolomics and lipidomics, provides hope that complex molecular biomarker profiles could be developed to diagnose PsA. Importantly, the integration of these markers with high-throughput histology, imaging and standardized clinical assessment data provides an important opportunity to develop molecular profiles that could improve the diagnosis of PsA, predict its occurrence in cohorts of individuals with psoriasis, differentiate PsA from other IMIDs, and improve therapeutic responses. In this review, we consider the technologies that are currently deployed in the EU IMI2 project HIPPOCRATES to define biomarker profiles specific for PsA and discuss the advantages of combining multi-omics data to improve the outcome of PsA patients.
Collapse
Affiliation(s)
- Robert Gurke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Annika Bendes
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - John Bowes
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WU, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Michaela Koehm
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | | | - Anne Barton
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WU, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Dirk Elewaut
- VIB-UGent Center for Inflammation Research, Ghent University, 9052 Ghent, Belgium
| | - Carl Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8QQ, UK
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | - Ewan Hunter
- Oxford BioDynamics Limited, Oxford OX4 2JZ, UK
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Vassilios Ioannidis
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Sabine Kugler
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer IAIS, Institute for Intelligent Analysis and Information Systems, Schloss Birlinghoven 1, 53757 Sankt Augustin, Germany
| | - Rik J. Lories
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, P.O. Box 813 O&N, Herestraat 49, 3000 Leuven, Belgium
| | - Eduard Resch
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Stefan Rüping
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer IAIS, Institute for Intelligent Analysis and Information Systems, Schloss Birlinghoven 1, 53757 Sankt Augustin, Germany
| | - Klaus Scholich
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jochen M. Schwenk
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - James C. Waddington
- Atturos Ltd., c/o UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Phil Whitfield
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, Garscube Campus, University of Glasgow, Glasgow G61 1QH, UK
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Oliver FitzGerald
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Frank Behrens
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Stephen R. Pennington
- Atturos Ltd., c/o UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | | |
Collapse
|
12
|
Sangouni AA, Baghban F, Khosravi M, Mozaffari-Khosravi H, Dehghan A, Hosseinzadeh M. Effect of L-carnitine supplementation on lipid accumulation product and cardiovascular indices in women with overweight/obesity who have knee osteoarthritis: a randomized controlled trial. BMC Rheumatol 2022; 6:53. [PMID: 36131324 PMCID: PMC9494899 DOI: 10.1186/s41927-022-00286-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Background Osteoarthritis is associated with obesity, dyslipidemia and cardiovascular diseases. It has been hypothesized that L-carnitine can improve cardiovascular risk factors. We aimed to investigate the effect of L-carnitine supplementation on lipid accumulation product (LAP) and atherogenic indices in women with overweight/obesity who have knee osteoarthritis. Methods In this double-blind randomized controlled trial, seventy-six women with overweight/obesity who had knee osteoarthritis were assigned into the intervention group and control group for 12 weeks. The intervention group received 1000 mg/day L-carnitine as capsule, and the control group received placebo. The primary outcomes were LAP, atherogenic index of plasma (AIP), atherogenic coefficient (AC) and Castelli risk index II (CRI-II). Results We found no significant difference between the groups in baseline values of LAP, AIP, AC and CRI-II. After the intervention, a significant reduction in LAP was observed in intervention group compared to the control group (− 11.05 (− 28.24 to 0.40) vs. − 5.82 (− 24.44 to 2.68); P = 0.03). However, there was no significant difference between two groups in AIP (− 0.05 ± 0.16 vs. − 0.01 ± 0.13; P = 0.19), AC (− 0.40 ± 0.81 vs. − 0.30 ± 0.67; P = 0.67) and CRI-II (− 0.20 ± 0.76 vs. − 0.21 ± 0.47; P = 0.11). Conclusions L-carnitine supplementation for 12 weeks can improve LAP, but it has no effect on cardiovascular outcomes. To reach a definitive conclusion, further clinical trials with larger sample sizes and higher dosages of L-carnitine are needed. Trial registration Registered on 27/4/2017 at Iranian Registry of Clinical Trials IRCT2017011932026N2.
Collapse
Affiliation(s)
- Abbas Ali Sangouni
- Nutrition and Food Security Research Center, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farnaz Baghban
- Nutrition and Food Security Research Center, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maryam Khosravi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hassan Mozaffari-Khosravi
- Nutrition and Food Security Research Center, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Dehghan
- Department of Internal Medicine, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Hosseinzadeh
- Nutrition and Food Security Research Center, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. .,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
13
|
Halim NFAA, Ali MSM, Leow ATC, Rahman RNZRA. Membrane fatty acid desaturase: biosynthesis, mechanism, and architecture. Appl Microbiol Biotechnol 2022; 106:5957-5972. [PMID: 36063178 DOI: 10.1007/s00253-022-12142-3] [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: 12/29/2021] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022]
Abstract
Fatty acid desaturase catalyzes the desaturation reactions by inserting double bonds into the fatty acyl chain, producing unsaturated fatty acids, which play a vital part in the synthesis of polyunsaturated fatty acids. Though soluble fatty acid desaturases have been described extensively in advanced organisms, there are very limited studies of membrane fatty acid desaturases due to their difficulties in producing a sufficient amount of recombinant desaturases. However, the advancement of technology has shown substantial progress towards the development of elucidating crystal structures of membrane fatty acid desaturase, thus, allowing modification of structure to be manipulated. Understanding the structure, mechanism, and biosynthesis of fatty acid desaturase lay a foundation for the potential production of various strategies associated with alteration and modifications of polyunsaturated fatty acids. This manuscript presents the current state of knowledge and understanding about the structure, mechanisms, and biosynthesis of fatty acid desaturase. In addition, the role of unsaturated fatty acid desaturases in health and diseases is also encompassed. This will be useful in understanding the molecular basis and structural protein of fatty acid desaturase that are significant for the advancement of therapeutic strategies associated with the improvement of health status. KEY POINTS: • Current state of knowledge and understanding about the biosynthesis, mechanisms, and structure of fatty acid desaturase. • The role of unsaturated fatty acid desaturase. • The molecular basis and structural protein elucidated the crystal structure of fatty acid desaturase.
Collapse
Affiliation(s)
- Nur Farah Anis Abd Halim
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mohd Shukuri Mohamad Ali
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Adam Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Raja Noor Zaliha Raja Abd Rahman
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
14
|
Moore L, Pan Z, Brotto M. RNAseq of Osteoarthritic Synovial Tissues: Systematic Literary Review. FRONTIERS IN AGING 2022; 3:836791. [PMID: 35821799 PMCID: PMC9261452 DOI: 10.3389/fragi.2022.836791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022]
Abstract
Osteoarthritis (OA) is one of the most common causes of disability in aged people, and it is defined as a degenerative arthropathy, characterized by the disruption in joint tissue. The synovium plays a vital role in maintaining the health of the joint by supplying the nutrients to the surrounding tissues and the lubrication for joint movement. While it is well known that all the joint tissues are communicating and working together to provide a functioning joint, most studies on OA have been focused on bone and cartilage but much less about synovium have been reported. The purpose of this review was to investigate the current literature focused on RNA sequencing (RNAseq) of osteoarthritic synovial tissues to further understand the dynamic transcriptome changes occurring in this pivotal joint tissue. A total of 3 electronic databases (PubMed, CINHAL Complete, and Academic Complete) were systematically searched following PRISMA guidelines. The following criteria was used for inclusion: English language, free full text, between the period 2011–2022, size of sample (n > 10), study design being either retrospective or prospective, and RNAseq data of synovial tissue from OA subjects. From the initial search, 174 articles, 5 met all of our criteria and were selected for this review. The RNAseq analysis revealed several differentially expressed genes (DEGs) in synovial tissue. These genes are related to the inflammatory pathway and regulation of the extracellular matrix. The MMP family, particularly MMP13 was identified by three of the studies, indicating its important role in OA. IL6, a key contributor in the inflammation pathway, was also identified in 3 studies. There was a total of 8 DEGs, MMP13, MMP1, MMP2, APOD, IL6, TNFAIP6, FCER1G, and IGF1 that overlapped in 4 out of the 5 studies. One study focused on microbial RNA in the synovial tissue found that the microbes were differentially expressed in OA subjects too. These differentially expressed microbes have also been linked to the inflammatory pathway. Further investigation with more clinical gene profiling in synovial tissue of OA subjects is required to reveal the causation and progression, as well as aid in the development of new treatments.
Collapse
|
15
|
Dogan S, Kimyon G, Ozkan H, Kacmaz F, Camdeviren B, Karaaslan I. TNF-alpha, IL-6, IL-10 and fatty acids in rheumatoid arthritis patients receiving cDMARD and bDMARD therapy. Clin Rheumatol 2022; 41:2341-2349. [PMID: 35467309 DOI: 10.1007/s10067-022-06180-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE The present study aimed to examine the effects of cDMARD and bDMARD therapy on both gene expressions and protein levels of TNF-α, IL-6, IL-10 and fatty acid levels in patients with RA. METHOD Plasma TNF-α, IL-6, and IL-10 levels were examined by the ELISA method, while TNF-α, IL-6, and IL-10 gene expression levels were examined by RT-qPCR, and fatty acid levels were examined by GC/MS. RESULTS IL-10 gene expression levels significantly increased in RA patients receiving cDMARD treatment compared to those of the control group. Also, eicosadienoic acid, myristoleic acid and capric acid levels were significantly lower in the patient groups compared to those in the control group. CONCLUSION The drugs used in the treatment of RA had no effect on the fatty acid levels whereas had effects on the mRNA and protein levels of the target cytokines.
Collapse
Affiliation(s)
- Serdar Dogan
- Department of Biochemistry, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey.
| | - Gezmis Kimyon
- Department of Rheumatology, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Huseyin Ozkan
- Department of Genetics, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Filiz Kacmaz
- Department of Molecular Biochemistry and Genetics, Health Science Institute, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Baran Camdeviren
- Department of Molecular Biochemistry and Genetics, Health Science Institute, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| | - Irem Karaaslan
- Research and Application Center for Technology and Research and Development, Hatay Mustafa Kemal University, Antakya, Hatay, 31060, Turkey
| |
Collapse
|
16
|
Cao X, Cui Z, Ding Z, Chen Y, Wu S, Wang X, Huang J. An osteoarthritis subtype characterized by synovial lipid metabolism disorder and fibroblast-like synoviocyte dysfunction. J Orthop Translat 2022; 33:142-152. [PMID: 35330945 PMCID: PMC8919236 DOI: 10.1016/j.jot.2022.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 11/25/2022] Open
Abstract
Background The heterogeneity of osteoarthritis (OA) significantly limits the effectiveness of pharmacological treatments in an unselected patient population. In this context, the identification of OA subtypes is meaningful for the development of therapies that target specific types of OA pathogenesis. Methods Expression array profiles of 70 OA and 36 control synovial samples were extracted from the GEO database. Unsupervised consensus clustering was performed based on the most variable genes to identify OA subclusters. Next, Joint samples from OA patients were obtained. We divided the OA patient into two subpopulations according to synovial ADCY7 levels. Synovium and cartilage samples from different OA subpopulations were evaluated. In addition, we established a high-fat diet (HFD)-induced rat OA model. We evaluated OA progression, lipid metabolism, synovitis and fibroblast-like synoviocytes (FLS) function in this HFD-induced OA model. Results 70 OA patients were categorized into three distinct subclusters. We noted that one subcluster was characterized by synovial lipid metabolism disorder GO terms. We further identified the most noticeable KEGG pathway “Regulation of lipolysis in adipocytes” in this subcluster as well as the most significantly differentially expressed gene, ADCY7. We found that the ADCY7 high expressing group (32.6%) exhibited features of synovial inflammatory lipolysis epithelial-mesenchymal transition (EMT) tendency, as well as faster join space narrowing. The HFD induced OA-like degeneration in rat joints. We observed similar synovial inflammatory lipolysis and EMT in FLS, characterized by higher proliferative and invasive activity and elevated proinflammatory and procatabolic properties. ADCY7 was highly expressed in the synovium of the HFD-OA model rats and the inhibition of ADCY7 effectively attenuated these HFD-induced degenerative changes as well as synovial inflammatory lipolysis and FLS dysfunction. In HFD-FLSs, ADCY7 promoted the phosphorylation of PKA as well as its downstream lipid droplet-associated protein PLIN1 and hormone-sensitive lipase (HSL). The inhibition of PKA largely alleviated ADCY7-mediated HFD-FLS dysfunction. Conclusions We described a synovial EMT and lipid metabolism disorder in the pathogenesis of OA. This novel mechanism may represent a currently undefined OA subtype. ADCY7 is a potential molecular marker of this pathomechanism. The Translational potential of this article Utilizing synovial samples from OA patients, we identified a subpopulation with high ADCY7 expression. This may represent a currently undefined OA subtype and explain the clinical phenomenon of more severe synovial inflammation in obese OA patients. In addition, we established an HFD-induced OA rat model and found an upregulation of ADCY7 in the synovium. We confirmed that the inhibition of ADCY7 could effectively attenuate HFD-induced degenerative changes as well as the inflammatory lipolysis and FLS dysfunction observed in the rat model. This suggests that ADCY7 and its downstream pathways are potential pharmacological targets for treating this lipid-metabolism-disorder-related OA mechanism.
Collapse
|
17
|
Lipidomic Profiling Identifies Serum Lipids Associated with Persistent Multisite Musculoskeletal Pain. Metabolites 2022; 12:metabo12030206. [PMID: 35323649 PMCID: PMC8953175 DOI: 10.3390/metabo12030206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 01/27/2023] Open
Abstract
Lipid mediators have been suggested to have a role in pain sensitivity and response; however, longitudinal data on lipid metabolites and persistent multisite musculoskeletal pain (MSMP) are lacking. This study was to identify lipid metabolic markers for persistent MSMP. Lipidomic profiling of 807 lipid species was performed on serum samples of 536 participants from a cohort study. MSMP was measured by a questionnaire and defined as painful sites ≥4. Persistent MSMP was defined as having MSMP at every visit. Logistic regression was used with adjustment for potential confounders. The Benjamini–Hochberg method was used to control for multiple testing. A total of 530 samples with 807 lipid metabolites passed quality control. Mean age at baseline was 61.54 ± 6.57 years and 50% were females. In total, 112 (21%) of the participants had persistent MSMP. Persistent MSMP was significantly associated with lower levels of monohexosylceramide (HexCer)(d18:1/22:0 and d18:1/24:0), acylcarnitine (AC)(26:0) and lysophosphatidylcholine (LPC)(18:1 [sn1], 18:2 [sn1], 18:2 [sn2], and 15-MHDA[sn1] [104_sn1]) after controlling for multiple testing. After adjustment for age, sex, body mass index, comorbidities, and physical activity, HexCer(d18:1/22:0 and d18:1/24:0) and LPC(15-MHDA [sn1] [104_sn1]) were significantly associated with persistent MSMP [Odds Ratio (OR) ranging from 0.25–0.36]. Two lipid classes—HexCer and LPC—were negatively associated with persistent MSMP after adjustment for covariates (OR = 0.22 and 0.27, respectively). This study identified three novel lipid signatures of persistent MSMP, suggesting that lipid metabolism is involved in the pathogenesis of persistent pain.
Collapse
|
18
|
Huq M, Rosales-Solano H, Pawliszyn J. Investigation of binding of fatty acids to serum albumin to determine free concentrations: Experimental and in-silico approaches. Anal Chim Acta 2022; 1192:339370. [DOI: 10.1016/j.aca.2021.339370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/17/2022]
|
19
|
Metabolomics of Synovial Fluid and Infrapatellar Fat Pad in Patients with Osteoarthritis or Rheumatoid Arthritis. Inflammation 2022; 45:1101-1117. [PMID: 35041143 PMCID: PMC9095531 DOI: 10.1007/s10753-021-01604-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) and autoimmune-driven rheumatoid arthritis (RA) are inflammatory joint diseases with complex and insufficiently understood pathogeneses. Our objective was to characterize the metabolic fingerprints of synovial fluid (SF) and its adjacent infrapatellar fat pad (IFP) obtained during the same surgical operation from OA and RA knees. Non-targeted metabolite profiling was performed for 5 non-inflammatory trauma controls, 10 primary OA (pOA) patients, and 10 seropositive RA patients with high-resolution mass spectrometry-based techniques, and metabolites were matched with known metabolite identities. Groupwise differences in metabolic features were analyzed with the univariate Welch’s t-test and the multivariate linear discriminant analysis (LDA) and principal component analysis (PCA). Significant discrimination of metabolite profiles was discovered by LDA for both SF and IFP and by PCA for SF based on diagnosis. In addition to a few drug-derived substances, there were 16 and 13 identified metabolites with significant differences between the diagnoses in SF and IFP, respectively. The pathways downregulated in RA included androgen, bile acid, amino acid, and histamine metabolism, and those upregulated included biotin metabolism in pOA and purine metabolism in RA and pOA. The RA-induced downregulation of androgen and bile acid metabolism was observed for both SF and IFP. The levels of 11 lipid metabolites, mostly glycerophospholipids and fatty acid amides, were also altered by these inflammatory conditions. The identified metabolic pathways could be utilized in the future to deepen our understanding of the pathogeneses of OA and RA and to develop not only biomarkers for their early diagnosis but also therapeutic targets.
Collapse
|
20
|
Afzali MF, Radakovich LB, Sykes MM, Campbell MA, Patton KM, Sanford JL, Vigon N, Ek R, Narez GE, Marolf AJ, Sikes KJ, Haut Donahue TL, Santangelo KS. Early removal of the infrapatellar fat pad/synovium complex beneficially alters the pathogenesis of moderate stage idiopathic knee osteoarthritis in male Dunkin Hartley guinea pigs. Arthritis Res Ther 2022; 24:282. [PMID: 36578046 PMCID: PMC9795160 DOI: 10.1186/s13075-022-02971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The infrapatellar fat pad (IFP) is the largest adipose deposit in the knee; however, its contributions to the homeostasis of this organ remain undefined. To determine the influence of the IFP and its associated synovium (IFP/synovium complex or IFP/SC) on joint health, this study evaluated the progression of osteoarthritis (OA) following excision of this unit in a rodent model of naturally-occurring disease. METHODS Male Dunkin-Hartley guinea pigs (n=18) received surgical removal of the IFP in one knee at 3 months of age; contralateral knees received sham surgery as matched internal controls. Mobility and gait assessments were performed prior to IFP/SC removal and monthly thereafter. Animals were harvested at 7 months of age. Ten set of these knees were processed for microcomputed tomography (microCT), histopathology, transcript expression analyses, and immunohistochemistry (IHC); 8 sets of knees were dedicated to microCT and biomechanical testing (material properties of knee joints tissues and anterior drawer laxity). RESULTS Fibrous connective tissue (FCT) developed in place of the native adipose depot. Gait demonstrated no significant differences between IFP/SC removal and contralateral hindlimbs. MicroCT OA scores were improved in knees containing the FCT. Quantitatively, IFP/SC-containing knees had more osteophyte development and increased trabecular volume bone mineral density (vBMD) in femora and tibiae. Histopathology confirmed maintenance of articular cartilage structure, proteoglycan content, and chondrocyte cellularity in FCT-containing knees. Transcript analyses revealed decreased expression of adipose-related molecules and select inflammatory mediators in FCTs compared to IFP/SCs. This was verified via IHC for two key inflammatory agents. The medial articular cartilage in knees with native IFP/SCs showed an increase in equilibrium modulus, which correlated with increased amounts of magnesium and phosphorus. DISCUSSION/CONCLUSION Formation of the FCT resulted in reduced OA-associated changes in both bone and cartilage. This benefit may be associated with: a decrease in inflammatory mediators at transcript and protein levels; and/or improved biomechanical properties. Thus, the IFP/SC may play a role in the pathogenesis of knee OA in this strain, with removal prior to disease onset appearing to have short-term benefits.
Collapse
Affiliation(s)
- Maryam F. Afzali
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| | - Lauren B. Radakovich
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| | - Madeline M. Sykes
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| | - Margaret A. Campbell
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| | - Kayley M. Patton
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| | - Joseph L. Sanford
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| | - Nicole Vigon
- grid.266683.f0000 0001 2166 5835Department of Biomedical Engineering, S631 Life Sciences Laboratory, University of Massachusetts, Amherst, 240 Thatcher Road, Amherst, MA 01003 USA
| | - Ryan Ek
- grid.266683.f0000 0001 2166 5835Department of Biomedical Engineering, S631 Life Sciences Laboratory, University of Massachusetts, Amherst, 240 Thatcher Road, Amherst, MA 01003 USA
| | - Gerardo E. Narez
- grid.266683.f0000 0001 2166 5835Department of Biomedical Engineering, S631 Life Sciences Laboratory, University of Massachusetts, Amherst, 240 Thatcher Road, Amherst, MA 01003 USA
| | - Angela J. Marolf
- grid.47894.360000 0004 1936 8083Department of Environmental and Radiological Health Sciences, Colorado State University, 123 Flint Cancer Center, Fort Collins, CO 80523 USA
| | - Katie J. Sikes
- grid.47894.360000 0004 1936 8083Department of Clinical Sciences, Colorado State University, 1678 Clinical Sciences, Fort Collins, CO 80523 USA
| | - Tammy L. Haut Donahue
- grid.56061.340000 0000 9560 654XBiomedical Engineering Department, The University of Memphis, 3806 Norriswood, Memphis, TN 38152 USA
| | - Kelly S. Santangelo
- grid.47894.360000 0004 1936 8083Department of Microbiology, Immunology and Pathology, Colorado State University, 200 West Lake Street, Fort Collins, CO 80523 USA
| |
Collapse
|
21
|
Wang Z, Liu M, Zhou Y, Shao H, Yang D, Huang Y, Deng W. Effect of blood lipids and lipid-lowering therapies on osteoarthritis risk: A Mendelian randomization study. Front Med (Lausanne) 2022; 9:990569. [PMID: 36438033 PMCID: PMC9691771 DOI: 10.3389/fmed.2022.990569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background We aimed to investigate the effects of blood lipids and lipid-lowering agents on osteoarthritis (OA) risk. Materials and methods We performed Mendelian randomization (MR) analyses to estimate the causal effect of blood low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglyceride (TG) levels on knee and hip OA. Single nucleotide polymorphisms (SNPs) were selected from large genome-wide association studies (GWASs) of individuals of European ancestry as genetic instruments for blood lipid levels. The associations of selected genetic instruments with knee and hip OA were estimated in a recent GWAS of the UK Biobank and arcOGEN datasets. Univariate and multivariate MR analyses were performed to detect and adjust for potential pleiotropy. Furthermore, genetic instruments in HMGCR, NPC1L1, and PCSK9 regions were used to mimic LDL-C-lowering effects of statin, ezetimibe, and evolocumab, respectively. Results Genetically determined LDL-C increments led to reduced risks of both knee OA (OR = 0.91 per 1-SD increment, 95% CI: 0.86-0.95, P = 6.3 × 10-5) and hip OA (OR = 0.92, 95% CI: 0.85-0.99, P = 0.027). Multivariate MR analysis proved that the effect was independent of HDL-C, TG, and body mass index. TG increment was associated with reduced risks of hip OA in the univariate MR analysis; however, this was not supported by the multivariate MR analysis. Genetically proxied LDL-C-lowering effects of statins are related to increased risks of knee OA but not hip OA. Conclusions The findings suggested that LDL-C increments have independent protective effects on both knee and hip OA. LDL-C-lowering effects of statins may increase the risk of knee OA.
Collapse
Affiliation(s)
- Zhaolun Wang
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Mengyuan Liu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixin Zhou
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Hongyi Shao
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Dejin Yang
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Yong Huang
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Wang Deng
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| |
Collapse
|
22
|
Luan H, Gu W, Li H, Wang Z, Lu L, Ke M, Lu J, Chen W, Lan Z, Xiao Y, Xu J, Zhang Y, Cai Z, Liu S, Zhang W. Serum metabolomic and lipidomic profiling identifies diagnostic biomarkers for seropositive and seronegative rheumatoid arthritis patients. J Transl Med 2021; 19:500. [PMID: 34876179 PMCID: PMC8650414 DOI: 10.1186/s12967-021-03169-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Diagnosing seronegative rheumatoid arthritis (RA) can be challenging due to complex diagnostic criteria. We sought to discover diagnostic biomarkers for seronegative RA cases by studying metabolomic and lipidomic changes in RA patient serum. METHODS We performed comprehensive metabolomic and lipidomic profiling in serum of 225 RA patients and 100 normal controls. These samples were divided into a discovery set (n = 243) and a validation set (n = 82). A machine-learning-based multivariate classification model was constructed using distinctive metabolites and lipids signals. RESULTS Twenty-six metabolites and lipids were identified from the discovery cohort to construct a RA diagnosis model. The model was subsequently tested on a validation set and achieved accuracy of 90.2%, with sensitivity of 89.7% and specificity of 90.6%. Both seropositive and seronegative patients were identified using this model. A co-occurrence network using serum omics profiles was built and parsed into six modules, showing significant association between the inflammation and immune activity markers and aberrant metabolism of energy metabolism, lipids metabolism and amino acid metabolism. Acyl carnitines (20:3), aspartyl-phenylalanine, pipecolic acid, phosphatidylethanolamine PE (18:1) and lysophosphatidylethanolamine LPE (20:3) were positively correlated with the RA disease activity, while histidine and phosphatidic acid PA (28:0) were negatively correlated with the RA disease activity. CONCLUSIONS A panel of 26 serum markers were selected from omics profiles to build a machine-learning-based prediction model that could aid in diagnosing seronegative RA patients. Potential markers were also identified in stratifying RA cases based on disease activity.
Collapse
Affiliation(s)
- Hemi Luan
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China
| | - Wanjian Gu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Hua Li
- Sustech Core Research Facilities, Southern University of Science and Technology, Shenzhen, China
| | - Zi Wang
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China
| | - Lu Lu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Mengying Ke
- College of Pharmacy, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Jiawei Lu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenjun Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Zhangzhang Lan
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China
| | - Yanlin Xiao
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China
| | - Jinyue Xu
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China
| | - Yi Zhang
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis (SKLEBA), Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Shijia Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Wenyong Zhang
- School of Medicine, Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, 1088 Xueyuan Rd., Shenzhen, China.
| |
Collapse
|
23
|
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: 23] [Impact Index Per Article: 5.8] [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
|
24
|
Chen Y, Xu X, Li X, Zhong J, Wu B, Shang J, Jiang N, Wang B, Zhong S, Lu H. Identification of circular RNAs hsa_circ_0140271 in peripheral blood mononuclear cells as a novel diagnostic biomarker for female rheumatoid arthritis. J Orthop Surg Res 2021; 16:647. [PMID: 34717684 PMCID: PMC8557002 DOI: 10.1186/s13018-021-02794-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease, which commonly affects women. Accumulating evidence shows that differentially expressed circular RNAs (circRNAs) play crucial roles in the progress of RA. However, the roles of circRNAs in female RA remains unclear. This study explores potential role and diagnostic value of hsa_circ_0140271 from peripheral blood mononuclear cells (PBMC) in female RA. Methods Differential expression of circRNAs was determined by RNA-sequencing in PBMC from 4 healthy controls (HC) and 4 RA patients, and we further measured the level of hsa_circ_0140271 in a validation cohort consisting of 47 RA and 47 HC via RT-qPCR. Besides, correlation studies with clinical variables were also examined. What’s more, we performed bioinformatics analysis to predict the potential role of hsa_circ_0140271. Results PBMC expression of hsa_circ_0140271 of female RA was significantly higher than that of female HC, and it was positively correlated with antistreptolysin (ASO). Furthermore, the receiver operating characteristic (ROC) curve indicated that hsa_circ_0140271 could distinguish female RA from female HC and female patients with ankylosing spondylitis (AS) or osteoarthritis (OA). Besides, the combined diagnosis anti-cyclic citrullinated peptide (Anti-CCP) + hsa_circ_0140271 could improve diagnostic accuracy with an area under the curve (AUC) of 0.818 to compared with Anti-CCP. Furthermore, KEGG pathway enrichment analysis indicated hsa_circ_0140271 may act as microRNA sponge and participate in fatty acid metabolism pathways. Conclusion Hsa_circ_0140271 was likely to be used as a promising diagnostic biomarker for female RA; it may act as microRNA sponge to regulate fatty acid metabolism pathways in RA. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-021-02794-8.
Collapse
Affiliation(s)
- Yufeng Chen
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Xianghe Xu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Xuegang Li
- Department of Rheumatology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Junlong Zhong
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Biao Wu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Jie Shang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Ning Jiang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Bin Wang
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China
| | - Shuping Zhong
- Department of Rheumatology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.
| | - Huading Lu
- Department of Orthopaedics, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
25
|
Zhang Z, Liu N, Chen X, Zhang F, Kong T, Tang X, Yang Q, Chen W, Xiong X, Chen X. UCHL1 regulates inflammation via MAPK and NF-κB pathways in LPS-activated macrophages. Cell Biol Int 2021; 45:2107-2117. [PMID: 34288216 DOI: 10.1002/cbin.11662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 06/28/2021] [Accepted: 07/03/2021] [Indexed: 12/27/2022]
Abstract
Inflammation is a common pathophysiological process as well as a clinical threat that occurs in various diseases worldwide. It is well-documented that nuclear factor-κB (NF-κB) and mitogen-activated protein kinase pathways are involved in inflammatory reactions to microbial infections in lipopolysaccharide (LPS)-activated macrophages. The deubiquitinase ubiquitin carboxyl-terminal hydrolase-L1 (UCHL1) has been reported as an oncoprotein to promote the growth and progression of cancer cells. However, the regulatory mechanism of UCHL1 in inflammation is currently unclear. Here, we aimed to assess the effects of UCHL1 on LPS-associated inflammatory response in vitro and in vivo by enzyme-linked immunosorbent assay, quantitative reverse-transcription polymerase chain reaction, and western blot analysis. This study identified that inhibition or knockdown of UCHL1 decreased the amounts of the key pro-inflammatory cytokines, including interleukin-6 and tumor necrosis factor-α in macrophages. Additionally, inhibition of UCHL1 suppressed LPS-induced extracellular signal-regulated protein kinase 1/2 phosphorylation and NF-κB translocation by regulating the inhibitor of NF-κB. Mechanically, UCHL1 interacts with IκBα protein in THP-1. Meanwhile, inhibition of UCHL1 blocked the LPS-induced degradation of IκBα through the ubiquitin-proteasome system. Moreover, in vivo assay showed that suppression of UCHL1 notably reduced the LPS-induced animal death and release of pro-inflammatory cytokines. Overall, the current findings uncover that UCHL1 functions as a crucial regulator for inflammatory response via reversing the degradation of IκBα, representing a potential target for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Zhenhui Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohua Chen
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fangcheng Zhang
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tianyu Kong
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoyan Tang
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qilin Yang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weiyan Chen
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuming Xiong
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohui Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Janovits PM, Leiguez E, Portas V, Teixeira C. A Metalloproteinase Induces an Inflammatory Response in Preadipocytes with the Activation of COX Signalling Pathways and Participation of Endogenous Phospholipases A 2. Biomolecules 2021; 11:921. [PMID: 34206390 PMCID: PMC8301905 DOI: 10.3390/biom11070921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes that have been associated with the pathogenesis of inflammatory diseases and obesity. Adipose tissue in turn is an active endocrine organ capable of secreting a range of proinflammatory mediators with autocrine and paracrine properties, which contribute to the inflammation of adipose tissue and adjacent tissues. However, the potential inflammatory effects of MMPs in adipose tissue cells are still unknown. This study investigates the effects of BmooMPα-I, a single-domain snake venom metalloproteinase (SVMP), in activating an inflammatory response by 3T3-L1 preadipocytes in culture, focusing on prostaglandins (PGs), cytokines, and adipocytokines biosynthesis and mechanisms involved in prostaglandin E2 (PGE2) release. The results show that BmooMPα-I induced the release of PGE2, prostaglandin I2 (PGI2), monocyte chemoattractant protein-1 (MCP-1), and adiponectin by preadipocytes. BmooMPα-I-induced PGE2 biosynthesis was dependent on group-IIA-secreted phospholipase A2 (sPLA2-IIA), cytosolic phospholipase A2-α (cPLA2-α), and cyclooxygenase (COX)-1 and -2 pathways. Moreover, BmooMPα-I upregulated COX-2 protein expression but not microsomal prostaglandin E synthase-1 (mPGES-1) expression. In addition, we demonstrate that the enzymatic activity of BmooMPα-I is essential for the activation of prostanoid synthesis pathways in preadipocytes. These data highlight preadipocytes as important targets for metalloproteinases and provide new insights into the contribution of these enzymes to the inflammation of adipose tissue and tissues adjacent to it.
Collapse
Affiliation(s)
- Priscila Motta Janovits
- Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil;
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Elbio Leiguez
- Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil;
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Viviane Portas
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
- Laboratório de Desenvolvimento e Inovação, Instituto Butantan, São Paulo 05503-900, Brazil
| | - Catarina Teixeira
- Laboratório de Farmacologia, Instituto Butantan, São Paulo 05503-900, Brazil;
- Centre of Excellence in New Target Discovery (CENTD), Instituto Butantan, São Paulo 05503-900, Brazil;
| |
Collapse
|
27
|
Jacenik D, Bagüés A, López-Gómez L, López-Tofiño Y, Iriondo-DeHond A, Serra C, Banovcanová L, Gálvez-Robleño C, Fichna J, del Castillo MD, Uranga JA, Abalo R. Changes in Fatty Acid Dietary Profile Affect the Brain-Gut Axis Functions of Healthy Young Adult Rats in a Sex-Dependent Manner. Nutrients 2021; 13:1864. [PMID: 34070787 PMCID: PMC8228732 DOI: 10.3390/nu13061864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/15/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Dietary modifications, including those affecting dietary fat and its fatty acid (FA) composition, may be involved in the development of brain-gut axis disorders, with different manifestations in males and females. Our aim was to evaluate the impact of three purified diets with different FA composition on the brain-gut axis in rats of both sexes. Male and female Wistar rats fed a cereal-based standard diet from weaning were used. At young adult age (2-3 months old), animals were divided into three groups and treated each with a different refined diet for 6 weeks: a control group fed on AIN-93G diet containing 7% soy oil (SOY), and two groups fed on AIN-93G modified diets with 3.5% soy oil replaced by 3.5% coconut oil (COCO) or 3.5% evening primrose oil (EP). Different brain-gut axis parameters were evaluated during 4-6 weeks of dietary intervention. Compared with SOY diet (14% saturated FAs, and 58% polyunsaturated FAs), COCO diet (52.2% saturated FAs and 30% polyunsaturated FAs) produced no changes in brain functions and minor gastrointestinal modifications, whereas EP diet (11.1% saturated FAs and 70.56% polyunsaturated FAs) tended to decrease self-care behavior and colonic propulsion in males, and significantly increased exploratory behavior, accelerated gastrointestinal transit, and decreased cecum and fecal pellet density in females. Changes in FA composition, particularly an increase in ω-6 polyunsaturated FAs, seem to facilitate the development of brain-gut axis alterations in a sex-dependent manner, with a relatively higher risk in females.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Ana Bagüés
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC), URJC, 28922 Alcorcón, Spain
- Associated I+D+i Unit to the Institute of Medicinal Chemistry (IQM), Scientific Research Superior Council (CSIC), 28006 Madrid, Spain
| | - Laura López-Gómez
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), URJC, 28922 Alcorcón, Spain
| | - Yolanda López-Tofiño
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), URJC, 28922 Alcorcón, Spain
| | - Amaia Iriondo-DeHond
- Food Bioscience Group, Department of Bioactivity and Food Analysis, Instituto de Investigación en Ciencias de la Alimentación (CIAL) (CSIC-UAM), Calle Nicolás Cabrera, 9, 28049 Madrid, Spain; (A.I.-D.); (M.D.d.C.)
| | - Cristina Serra
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
| | - Laura Banovcanová
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
| | - Carlos Gálvez-Robleño
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), URJC, 28922 Alcorcón, Spain
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Maria Dolores del Castillo
- Food Bioscience Group, Department of Bioactivity and Food Analysis, Instituto de Investigación en Ciencias de la Alimentación (CIAL) (CSIC-UAM), Calle Nicolás Cabrera, 9, 28049 Madrid, Spain; (A.I.-D.); (M.D.d.C.)
| | - José Antonio Uranga
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), URJC, 28922 Alcorcón, Spain
| | - Raquel Abalo
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (A.B.); (L.L.-G.); (Y.L.-T.); (C.S.); (L.B.); (C.G.-R.); (J.A.U.)
- Associated I+D+i Unit to the Institute of Medicinal Chemistry (IQM), Scientific Research Superior Council (CSIC), 28006 Madrid, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), URJC, 28922 Alcorcón, Spain
- Working Group of Basic Sciences in Pain and Analgesia of the Spanish Pain Society (Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia de la Sociedad Española del Dolor), 28046 Madrid, Spain
| |
Collapse
|
28
|
Synovial Fluid Fatty Acid Profiles Are Differently Altered by Inflammatory Joint Pathologies in the Shoulder and Knee Joints. BIOLOGY 2021; 10:biology10050401. [PMID: 34064447 PMCID: PMC8147852 DOI: 10.3390/biology10050401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022]
Abstract
Anomalies of fatty acid (FA) metabolism characterize osteoarthritis (OA) and rheumatoid arthritis (RA) in the knee joint. No previous study has investigated the synovial fluid (SF) FA manifestations in these aging-related inflammatory diseases in the shoulder. The present experiment compared the FA alterations between the shoulder and knee joints in patients with end-stage OA or end-stage RA. SF samples were collected during glenohumeral or knee joint surgery from trauma controls and from OA and RA patients (n = 42). The FA composition of SF total lipids was analyzed by gas chromatography with flame ionization and mass spectrometric detection and compared across cohorts. The FA signatures of trauma controls were mostly uniform in both anatomical locations. RA shoulders were characterized by elevated percentages of 20:4n-6 and 22:6n-3 and with reduced proportions of 18:1n-9. The FA profiles of OA and RA knees were relatively uniform and displayed lower proportions of 18:2n-6, 22:6n-3 and total n-6 polyunsaturated FAs (PUFAs). The results indicate location- and disease-dependent differences in the SF FA composition. These alterations in FA profiles and their potential implications for the production of PUFA-derived lipid mediators may affect joint lubrication, synovial inflammation and pannus formation as well as cartilage and bone degradation and contribute to the pathogeneses of inflammatory joint diseases.
Collapse
|
29
|
Rocha B, Cillero-Pastor B, Ruiz-Romero C, Paine MRL, Cañete JD, Heeren RMA, Blanco FJ. Identification of a distinct lipidomic profile in the osteoarthritic synovial membrane by mass spectrometry imaging. Osteoarthritis Cartilage 2021; 29:750-761. [PMID: 33582239 DOI: 10.1016/j.joca.2020.12.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/12/2020] [Accepted: 12/16/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Synovial inflammation is one of the most characteristic events in different types of arthritis, including Osteoarthritis (OA). Emerging evidence also suggests the involvement of lipids in the regulation of inflammatory processes. The aim of this study was to elucidate the heterogeneity and spatial distribution of lipids in the OA synovial membrane and explore their putative involvement in inflammation. METHOD The abundance and distribution of lipids were examined in human synovial membranes. To this end, histological cuts from this tissue were analysed by matrix-assisted laser desorption ionization - mass spectrometry imaging (MALDI-MSI). The lipidomic profile of OA synovium was characterized and compared with healthy and other forms of inflammatory arthropathies as Rheumatoid Arthritis (RA) and Psoriatic Arthritis (PsA) using principal component analysis and discriminant analysis methods. Lipid identification was undertaken by tandem MS analyses and database queries. RESULTS Our results reveal differential and characteristic lipidomic profiles between OA and control samples. Specifically, we unveiled that OA synovium presents elevated levels of phosphatidylcholines, fatty acids and lysophosphatidic acids and lower levels of lysophosphatidylcholines compared to control tissues. The spatial distribution of particular glycerophospholipids was also correlated with hypertrophic, inflamed or vascularized synovial areas. Compared with other inflammatory arthritis, the OA tissue showed lower amounts of phosphatidylethanolamine-based plasmalogens. CONCLUSIONS This study provides a novel insight into the lipid profiles of synovial membrane and differences in abundance between OA and control tissues. The lipidomic alterations improves understanding of the pathogenic mechanisms of OA and may be important for its diagnosis.
Collapse
Affiliation(s)
- B Rocha
- Grupo de Unidad de Proteómica, Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), C/ As Xubias de Arriba 84, 15006, A Coruña, Spain
| | - B Cillero-Pastor
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, the Netherlands
| | - C Ruiz-Romero
- Grupo de Unidad de Proteómica, Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), C/ As Xubias de Arriba 84, 15006, A Coruña, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11, 28029, Madrid, Spain.
| | - M R L Paine
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, the Netherlands
| | - J D Cañete
- Unidad de Artritis. Servicio de Reumatología. Hospital Clínico de Barcelona, Barcelona, Spain
| | - R M A Heeren
- The Maastricht Multimodal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, the Netherlands
| | - F J Blanco
- Grupo de Unidad de Proteómica, Grupo de Investigación de Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), C/ As Xubias de Arriba 84, 15006, A Coruña, Spain; Universidade da Coruña (UDC), Grupo de Investigación de Reumatología y Salud (GIR-S), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, 15008, A Coruña, Spain.
| |
Collapse
|
30
|
Fatty Acids and Oxylipins in Osteoarthritis and Rheumatoid Arthritis-a Complex Field with Significant Potential for Future Treatments. Curr Rheumatol Rep 2021; 23:41. [PMID: 33913032 PMCID: PMC8081702 DOI: 10.1007/s11926-021-01007-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Purpose of Review Osteoarthritis (OA) and rheumatoid arthritis (RA) are characterized by abnormal lipid metabolism manifested as altered fatty acid (FA) profiles of synovial fluid and tissues and in the way dietary FA supplements can influence the symptoms of especially RA. In addition to classic eicosanoids, the potential roles of polyunsaturated FA (PUFA)-derived specialized pro-resolving lipid mediators (SPM) have become the focus of intensive research. Here, we summarize the current state of knowledge of the roles of FA and oxylipins in the degradation or protection of synovial joints. Recent Findings There exists discordance between the large body of literature from cell culture and animal experiments on the adverse and beneficial effects of individual FA and the lack of effective treatments for joint destruction in OA and RA patients. Saturated 16:0 and 18:0 induce mostly deleterious effects, while long-chain n-3 PUFA, especially 20:5n-3, have positive influence on joint health. The situation can be more complex for n-6 PUFA, such as 18:2n-6, 20:4n-6, and its derivative prostaglandin E2, with a combination of potentially adverse and beneficial effects. SPM analogs have future potential as analgesics for arthritic pain. Summary Alterations in FA profiles and their potential implications in SPM production may affect joint lubrication, synovial inflammation, pannus formation, as well as cartilage and bone degradation and contribute to the pathogeneses of inflammatory joint diseases. Further research directions include high-quality randomized controlled trials on dietary FA supplements and investigations on the significance of lipid composition of microvesicle membrane and cargo in joint diseases.
Collapse
|
31
|
Effect of n-3 PUFA on extracellular matrix protein turnover in patients with psoriatic arthritis: a randomized, double-blind, placebo-controlled trial. Rheumatol Int 2021; 41:1065-1077. [PMID: 33885930 PMCID: PMC8079340 DOI: 10.1007/s00296-021-04861-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/07/2021] [Indexed: 12/03/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease characterized by involvement of skin, axial and peripheral skeleton. An altered balance between extracellular matrix (ECM) formation and breakdown is a key event in PsA, and changes in ECM protein metabolites may provide insight to tissue changes. Dietary fish oils (n-3 PUFA) might affect the inflammation driven tissue turnover. The aim was to evaluate ECM metabolites in patients with PsA compared to healthy individuals and investigate the effects of n-3 PUFA. The 24-week randomized, double-blind, placebo-controlled trial of PUFA included 142 patients with PsA. Fifty-seven healthy individuals were included for comparison. This study is a sub-study investigating biomarkers of tissue remodelling as secondary outcomes. Serum samples at baseline and 24 weeks and healthy individuals were obtained, while a panel of ECM metabolites reflecting bone and soft tissue turnover were measured by ELISAs: PRO-C1, PRO-C3, PRO-C4, C1M, C3M, C4M, CTX-I and Osteocalcin (OC). C1M, PRO-C3, PRO-C4 and C4M was found to be elevated in PsA patients compared to the healthy individuals (from 56 to 792%, all p < 0.0001), where no differences were found for OC, CTX-I, PRO-C1 and C3M. PRO-C3 was increased by 7% in patients receiving n-3 PUFA after 24 weeks compared to baseline levels (p = 0.002). None of the other biomarkers was changed with n-3 PUFA treatment. This indicates that tissue turnover is increased in PsA patients compared to healthy individuals, while n-3 PUFA treatment for 24 weeks did not have an effect on tissue turnover. Trial registration NCT01818804. Registered 27 March 2013–Completed 18 February 2016. https://clinicaltrials.gov/ct2/show/NCT01818804?term=NCT01818804&rank=1
Collapse
|
32
|
Higher PGD 2 production by synovial mast cells from rheumatoid arthritis patients compared with osteoarthritis patients via miR-199a-3p/prostaglandin synthetase 2 axis. Sci Rep 2021; 11:5738. [PMID: 33707464 PMCID: PMC7952410 DOI: 10.1038/s41598-021-84963-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
We previously reported that synovial mast cells (MCs) from patients with rheumatoid arthritis (RA) produced TNF-α in response to immune complexes via FcγRI and FcγRIIA. However, the specific functions of synovial MCs in RA remain unclear. This study aimed to elucidate those functions. Synovial tissues and fluid were obtained from RA and osteoarthritis (OA) patients undergoing joint replacement surgery. Synovium-derived, cultured MCs were generated by culturing dispersed synovial cells with stem cell factor. We performed microarray-based screening of mRNA and microRNA (miRNA), followed by quantitative RT-PCR-based verification. Synovial MCs from RA patients showed significantly higher prostaglandin systhetase (PTGS)1 and PTGS2 expression compared with OA patients' MCs, and they produced significantly more prostaglandin D2 (PGD2) following aggregation of FcγRI. PGD2 induced IL-8 production by human group 2 innate lymphoid cells, suggesting that PGD2-producing MCs induce neutrophil recruitment into the synovium of RA patients. PTGS2 mRNA expression in RA patients' MCs correlated inversely with miRNA-199a-3p expression, which down-regulated PTGS2. RA patients' synovial fluid contained significantly more PGD2 compared with OA patients' fluid. Synovial MCs might regulate inflammation in RA through hyper-production of PGD2 following FcRγ aggregation. Our findings indicate functional heterogeneity of human MCs among diseases.
Collapse
|
33
|
Zheng L, Zhang Z, Sheng P, Mobasheri A. The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res Rev 2021; 66:101249. [PMID: 33383189 DOI: 10.1016/j.arr.2020.101249] [Citation(s) in RCA: 345] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by low-grade inflammation and high levels of clinical heterogeneity. Aberrant chondrocyte metabolism is a response to changes in the inflammatory microenvironment and may play a key role in cartilage degeneration and OA progression. Under conditions of environmental stress, chondrocytes tend to adapt their metabolism to microenvironmental changes by shifting from one metabolic pathway to another, for example from oxidative phosphorylation to glycolysis. Similar changes occur in other joint cells, including synoviocytes. Switching between these pathways is implicated in metabolic alterations that involve mitochondrial dysfunction, enhanced anaerobic glycolysis, and altered lipid and amino acid metabolism. The shift between oxidative phosphorylation and glycolysis is mainly regulated by the AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) pathways. Chondrocyte metabolic changes are likely to be a feature of different OA phenotypes. Determining the role of chondrocyte metabolism in OA has revealed key features of disease pathogenesis. Future research should place greater emphasis on immunometabolism and altered metabolic pathways as a means to understand the pathophysiology of age-related OA. This knowledge will advance the development of new drugs against therapeutic targets of metabolic significance.
Collapse
Affiliation(s)
- Linli Zheng
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China
| | - Ziji Zhang
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China
| | - Puyi Sheng
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China.
| | - Ali Mobasheri
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080 China; Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, PO Box 5000, FI-90014 Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406, Vilnius, Lithuania; Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508 GA, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Casati S, Giannasi C, Niada S, Bergamaschi RF, Orioli M, Brini AT. Bioactive Lipids in MSCs Biology: State of the Art and Role in Inflammation. Int J Mol Sci 2021; 22:1481. [PMID: 33540695 PMCID: PMC7867257 DOI: 10.3390/ijms22031481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Lipidomics is a lipid-targeted metabolomics approach that aims to the comprehensive analysis of lipids in biological systems in order to highlight the specific functions of lipid species in health and disease. Lipids play pivotal roles as they are major structural components of the cellular membranes and energy storage molecules but also, as most recently shown, they act as functional and regulatory components of intra- and intercellular signaling. Herein, emphasis is given to the recently highlighted roles of specific bioactive lipids species, as polyunsaturated fatty acids (PUFA)-derived mediators (generally known as eicosanoids), endocannabinoids (eCBs), and lysophospholipids (LPLs), and their involvement in the mesenchymal stem cells (MSCs)-related inflammatory scenario. Indeed, MSCs are a heterogenous population of multipotent cells that have attracted much attention for their potential in regulating inflammation, immunomodulatory capabilities, and reparative roles. The lipidomics of the inflammatory disease osteoarthritis (OA) and the influence of MSCs-derived lipids have also been addressed.
Collapse
Affiliation(s)
- Sara Casati
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
| | - Chiara Giannasi
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
| | | | - Roberta F. Bergamaschi
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
| | - Marica Orioli
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
| | - Anna T. Brini
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, 20133 Milan, Italy; (C.G.); (R.F.B.); (M.O.); (A.T.B.)
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
| |
Collapse
|
35
|
Behl T, Kaur I, Sehgal A, Zengin G, Brisc C, Brisc MC, Munteanu MA, Nistor-Cseppento DC, Bungau S. The Lipid Paradox as a Metabolic Checkpoint and Its Therapeutic Significance in Ameliorating the Associated Cardiovascular Risks in Rheumatoid Arthritis Patients. Int J Mol Sci 2020; 21:ijms21249505. [PMID: 33327502 PMCID: PMC7764917 DOI: 10.3390/ijms21249505] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023] Open
Abstract
While the most common manifestations associated with rheumatoid arthritis (RA) are synovial damage and inflammation, the systemic effects of this autoimmune disorder are life-threatening, and are prevalent in 0.5–1% of the population, mainly associated with cardiovascular disorders (CVDs). Such effects have been instigated by an altered lipid profile in RA patients, which has been reported to correlate with CV risks. Altered lipid paradox is related to inflammatory burden in RA patients. The review highlights general lipid pathways (exogenous and endogenous), along with the changes in different forms of lipids and lipoproteins in RA conditions, which further contribute to elevated risks of CVDs like ischemic heart disease, atherosclerosis, myocardial infarction etc. The authors provide a deep insight on altered levels of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) and triglycerides (TGs) in RA patients and their consequence on the cardiovascular health of the patient. This is followed by a detailed description of the impact of anti-rheumatoid therapy on the lipid profile in RA patients, comprising DMARDs, corticosteroids, anti-TNF agents, anti-IL-6 agents, JAK inhibitors and statins. Furthermore, this review elaborates on the prospects to be considered to optimize future investigation on management of RA and treatment therapies targeting altered lipid paradigms in patients.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.)
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.)
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, 42130 Konya, Turkey;
| | - Ciprian Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (M.C.B.); (M.A.M.)
| | - Mihaela Cristina Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (M.C.B.); (M.A.M.)
| | - Mihai Alexandru Munteanu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (C.B.); (M.C.B.); (M.A.M.)
| | - Delia Carmen Nistor-Cseppento
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| |
Collapse
|
36
|
Shoieb SM, El-Ghiaty MA, El-Kadi AOS. Targeting arachidonic acid-related metabolites in COVID-19 patients: potential use of drug-loaded nanoparticles. EMERGENT MATERIALS 2020; 4:265-277. [PMID: 33225219 PMCID: PMC7670111 DOI: 10.1007/s42247-020-00136-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/23/2020] [Indexed: 05/02/2023]
Abstract
In March 2020, The World Health Organization (WHO) has declared that the coronavirus disease 2019 (COVID-19) is characterized as a global pandemic. As of September 2020, infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread to 213 countries and territories around the world, affected more than 31.5 million people, and caused more than 970,000 deaths worldwide. Although COVID-19 is a respiratory illness that mainly targets the lungs, it is currently well established that it is a multifactorial disease that affects other extra-pulmonary systems and strongly associated with a detrimental inflammatory response. Evidence has shown that SARS-CoV-2 causes perturbation in the arachidonic acid (AA) metabolic pathways; this disruption could lead to an imbalance between the pro-inflammatory metabolites of AA including mid-chain HETEs and terminal HETE (20-HETE) and the anti-inflammatory metabolites such as EETs and subterminal HETEs. Therefore, we propose novel therapeutic strategies to modulate the level of endogenous anti-inflammatory metabolites of AA and induce the patient's endogenous resolution mechanisms that will ameliorate the virus-associated systemic inflammation and enhance the primary outcomes in COVID-19 patients. Also, we propose that using nanoencapsulation of AA and its associated metabolites will contribute to the development of safer and more efficacious treatments for the management of COVID-19.
Collapse
Affiliation(s)
- Sherif M. Shoieb
- Faculty of Pharmacy & Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Alberta T6G 2E1 Canada
| | - Mahmoud A. El-Ghiaty
- Faculty of Pharmacy & Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Alberta T6G 2E1 Canada
| | - Ayman O. S. El-Kadi
- Faculty of Pharmacy & Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Alberta T6G 2E1 Canada
| |
Collapse
|
37
|
Norel X, Sugimoto Y, Ozen G, Abdelazeem H, Amgoud Y, Bouhadoun A, Bassiouni W, Goepp M, Mani S, Manikpurage HD, Senbel A, Longrois D, Heinemann A, Yao C, Clapp LH. International Union of Basic and Clinical Pharmacology. CIX. Differences and Similarities between Human and Rodent Prostaglandin E 2 Receptors (EP1-4) and Prostacyclin Receptor (IP): Specific Roles in Pathophysiologic Conditions. Pharmacol Rev 2020; 72:910-968. [PMID: 32962984 PMCID: PMC7509579 DOI: 10.1124/pr.120.019331] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Prostaglandins are derived from arachidonic acid metabolism through cyclooxygenase activities. Among prostaglandins (PGs), prostacyclin (PGI2) and PGE2 are strongly involved in the regulation of homeostasis and main physiologic functions. In addition, the synthesis of these two prostaglandins is significantly increased during inflammation. PGI2 and PGE2 exert their biologic actions by binding to their respective receptors, namely prostacyclin receptor (IP) and prostaglandin E2 receptor (EP) 1-4, which belong to the family of G-protein-coupled receptors. IP and EP1-4 receptors are widely distributed in the body and thus play various physiologic and pathophysiologic roles. In this review, we discuss the recent advances in studies using pharmacological approaches, genetically modified animals, and genome-wide association studies regarding the roles of IP and EP1-4 receptors in the immune, cardiovascular, nervous, gastrointestinal, respiratory, genitourinary, and musculoskeletal systems. In particular, we highlight similarities and differences between human and rodents in terms of the specific roles of IP and EP1-4 receptors and their downstream signaling pathways, functions, and activities for each biologic system. We also highlight the potential novel therapeutic benefit of targeting IP and EP1-4 receptors in several diseases based on the scientific advances, animal models, and human studies. SIGNIFICANCE STATEMENT: In this review, we present an update of the pathophysiologic role of the prostacyclin receptor, prostaglandin E2 receptor (EP) 1, EP2, EP3, and EP4 receptors when activated by the two main prostaglandins, namely prostacyclin and prostaglandin E2, produced during inflammatory conditions in human and rodents. In addition, this comparison of the published results in each tissue and/or pathology should facilitate the choice of the most appropriate model for the future studies.
Collapse
Affiliation(s)
- Xavier Norel
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Yukihiko Sugimoto
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Gulsev Ozen
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Heba Abdelazeem
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Yasmine Amgoud
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Amel Bouhadoun
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Wesam Bassiouni
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Marie Goepp
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Salma Mani
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Hasanga D Manikpurage
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Amira Senbel
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Dan Longrois
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Akos Heinemann
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Chengcan Yao
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| | - Lucie H Clapp
- Université de Paris, Institut National de la Sante et de la Recherche Medicale (INSERM), UMR-S 1148, CHU X. Bichat, Paris, France (X.N., G.O., H.A., Y.A., A.B., S.M., H.D.M., A.S., D.L.); Université Sorbonne Paris Nord, Villetaneuse, France (X.N., H.A., Y.A., A.B., S.M., D.L.); Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan (Y.S.); Istanbul University, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey (G.O.); Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.S., H.A., W.B.); Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom (C.Y., M.G.); Institut Supérieur de Biotechnologie de Monastir (ISBM), Université de Monastir, Monastir, Tunisia (S.M.); CHU X. Bichat, AP-HP, Paris, France (D.L.); Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria (A.H.); and Centre for Cardiovascular Physiology & Pharmacology, University College London, London, United Kingdom (L.H.C.)
| |
Collapse
|
38
|
Wang J, Dong J, Yang J, Wang Y, Liu J. Association between statin use and incidence or progression of osteoarthritis: meta-analysis of observational studies. Osteoarthritis Cartilage 2020; 28:1170-1179. [PMID: 32360737 DOI: 10.1016/j.joca.2020.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/26/2020] [Accepted: 04/21/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The objective of the study was to determine whether statin use could reduce the risk of the incidence or progression of osteoarthritis (OA). METHODS The PubMed, Embase, and Cochrane databases were systematically searched for observational studies on the association between statin use and OA. ORs and 95% CIs were directly retrieved or calculated. The Newcastle-Ottawa quality assessment scale was used for study quality assessment. Subgroup analysis, sensitivity analysis, and publication bias were conducted using Stata software. RESULTS A total of 11 studies (679807 participants) were identified from the systematic literature search. No significant association between statin use and incidence (OR = 1.010; 95% CI: 0.968 to 1.055; P = 0.638) or progression (OR = 1.076; 95% CI: 0.824 to 1.405; P = 0.589) of OA was found in our meta-analysis. The meta-analysis according to the symptomatic or radiological OA also found no significant association between statin use and OA. The subgroup analysis showed that atorvastatin (OR = 0.953; 95% CI: 0.911 to 0.998; P = 0.041) and rosuvastatin (OR = 1.180; 95% CI: 1.122 to 1.241; P < 0.0001) had opposite effects on OA. The results of the analysis according to the joint site, interval, and statin dose were all not significant. CONCLUSIONS Statin use may not be associated with a lower risk of incidence and progression of OA, regardless of joint site. The opposite effects of atorvastatin and rosuvastatin were detected in OA.
Collapse
Affiliation(s)
- J Wang
- Department of Orthopaedics, Tianjin Hospital, No. 406, Jiefang Nan Street, Hexi District, Tianjin, China.
| | - J Dong
- Department of Orthopaedics, Tianjin Hospital, No. 406, Jiefang Nan Street, Hexi District, Tianjin, China
| | - J Yang
- Department of Orthopaedics, Tianjin Hospital, No. 406, Jiefang Nan Street, Hexi District, Tianjin, China
| | - Y Wang
- Department of Orthopaedics, Tianjin Hospital, No. 406, Jiefang Nan Street, Hexi District, Tianjin, China
| | - J Liu
- Department of Orthopaedics, Tianjin Hospital, No. 406, Jiefang Nan Street, Hexi District, Tianjin, China
| |
Collapse
|
39
|
Greif DN, Kouroupis D, Murdock CJ, Griswold AJ, Kaplan LD, Best TM, Correa D. Infrapatellar Fat Pad/Synovium Complex in Early-Stage Knee Osteoarthritis: Potential New Target and Source of Therapeutic Mesenchymal Stem/Stromal Cells. Front Bioeng Biotechnol 2020; 8:860. [PMID: 32850724 PMCID: PMC7399076 DOI: 10.3389/fbioe.2020.00860] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
The infrapatellar fat pad (IFP) has until recently been viewed as a densely vascular and innervated intracapsular/extrasynovial tissue with biomechanical roles in the anterior compartment of the knee. Over the last decade, secondary to the proposition that the IFP and synovium function as a single unit, its recognized tight molecular crosstalk with emerging roles in the pathophysiology of joint disease, and the characterization of immune-related resident cells with varying phenotypes (e.g., pro and anti-inflammatory macrophages), this structural complex has gained increasing attention as a potential therapeutic target in patients with various knee pathologies including osteoarthritis (KOA). Furthermore, the description of the presence of mesenchymal stem/stromal cells (MSC) as perivascular cells within the IFP (IFP-MSC), exhibiting immunomodulatory, anti-fibrotic and neutralizing activities over key local mediators, has promoted the IFP as an alternative source of MSC for cell-based therapy protocols. These complementary concepts have supported the growing notion of immune and inflammatory events participating in the pathogenesis of KOA, with the IFP/synovium complex engaging not only in amplifying local pathological responses, but also as a reservoir of potential therapeutic cell-based products. Consequently, the aim of this review is to outline the latest discoveries related with the IFP/synovium complex as both an active participant during KOA initiation and progression thus emerging as a potential target, and a source of therapeutic IFP-MSCs. Finally, we discuss how these notions may help the design of novel treatments for KOA through modulation of local cellular and molecular cascades that ultimately lead to joint destruction.
Collapse
Affiliation(s)
- Dylan N Greif
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christopher J Murdock
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
40
|
Sano Y, Toyoshima S, Miki Y, Taketomi Y, Ito M, Lee H, Saito S, Murakami M, Okayama Y. Activation of inflammation and resolution pathways of lipid mediators in synovial fluid from patients with severe rheumatoid arthritis compared with severe osteoarthritis. Asia Pac Allergy 2020; 10:e21. [PMID: 32411586 PMCID: PMC7203435 DOI: 10.5415/apallergy.2020.10.e21] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/23/2020] [Indexed: 12/29/2022] Open
Abstract
Background The upregulation of the cyclooxygenase and lipoxygenase pathways of arachidonic acid is thought to be involved in the development of rheumatoid arthritis. Recently, the presence of specialized pro-resolving lipid mediators in synovial tissues from patients with osteoarthritis has been reported. Objective To clarify the quantitative and qualitative changes in lipid mediators in the synovium of severe rheumatoid arthritis patients, we compared the profiles of lipid mediators in synovial fluid obtained from patients with severe rheumatoid arthritis and from those with severe osteoarthritis. Methods We enrolled 18 patients with rheumatoid arthritis and 26 patients with osteoarthritis. All the patients had undergone total knee replacement surgery. Synovial fluid samples had been obtained during the surgery. Lipid profiling in the synovial fluid from these patients was performed using liquid chromatography-tandem mass spectrometry/mass spectrometry. Results Among the 150 oxidized fatty acids examined so far, 119 were substantially detected in synovial fluid from the patients. Not only the concentrations of pro-inflammatory lipid mediators such as prostaglandins and leukotrienes, but also those of specialized pro-resolving lipid mediators such as lipoxins, resolvins, and protectin D1 were significantly higher in synovial fluid obtained from rheumatoid arthritis patients than from synovial fluid obtained from osteoarthritis patients. Conclusion The activation of both inflammation and resolution pathways of lipid mediators might be a fatty acid signature in the synovial fluid of patients with severe rheumatoid arthritis. Inflammatory, anti-inflammatory and pro-resolving mediators in synovial fluid could be good biomarkers for differentiating between severe rheumatoid arthritis and severe osteoarthritis.
Collapse
Affiliation(s)
- Yutaka Sano
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan.,Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Shota Toyoshima
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan.,Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan.,Center for Medical Education, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Cellular Signaling, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mana Ito
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan.,Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Hyunho Lee
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Shu Saito
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Cellular Signaling, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Nihon University School of Medicine, Tokyo, Japan.,Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan.,Center for Medical Education, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
41
|
Zhang F, Xu R, Chai R, Xu Q, Liu M, Chen X, Chen X, Kong T, Zhang C, Liu SM, Zhang Z, Liu N. Deubiquitinase Inhibitor b-AP15 Attenuated LPS-Induced Inflammation via Inhibiting ERK1/2, JNK, and NF-Kappa B. Front Mol Biosci 2020; 7:49. [PMID: 32391376 PMCID: PMC7188916 DOI: 10.3389/fmolb.2020.00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/13/2020] [Indexed: 01/15/2023] Open
Abstract
b-AP15 is a deubiquitinase (DUB) inhibitor of 19S proteasomes, which in turn targets ubiquitin C-terminal hydrolase 5 (UCHL5) and ubiquitin-specific peptidase 14 (USP14). Nuclear factor kappa B (NF-κB) is closely linked to cellular response in macrophages when the organism is in the state of microbial infection, and it acts as a vital part in the mechanism of inflammatory reaction. However, the molecular mechanism by which DUB inhibitors, especially b-AP15, regulates inflammation remains poorly understood. This study aimed to investigate the relationship between b-AP15 and inflammation. The results showed that b-AP15 treatment significantly reduced the amounts of inflammatory indicators, such as tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in lipopolysaccharide (LPS)-stimulated THP-1 and macrophages. Meanwhile, similar results were obtained from in vivo experiments. In addition, b-AP15 also significantly improved the survival rate of sepsis mouse via high-density LPS mediation. Furthermore, b-AP15 also inhibited the ERK1/2 and JNK phosphorylation, increased IκBα levels, and inhibited NF-κB p65 by removing them from the cytoplasm into the nucleus. All these findings suggested that b-AP15 has anti-inflammatory action and acts as a potential neoteric target drug for treating microbial infection.
Collapse
Affiliation(s)
- Fangcheng Zhang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruqin Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Renjie Chai
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiong Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingke Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xuke Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaohua Chen
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tianyu Kong
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chongyu Zhang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shi-Ming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhenhui Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
42
|
Metabonomic-Transcriptome Integration Analysis on Osteoarthritis and Rheumatoid Arthritis. Int J Genomics 2020; 2020:5925126. [PMID: 31976312 PMCID: PMC6961787 DOI: 10.1155/2020/5925126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Purpose This study is aimed at exploring the potential metabolite/gene biomarkers, as well as the differences between the molecular mechanisms, of osteoarthritis (OA) and rheumatoid arthritis (RA). Methods Transcriptome dataset GSE100786 was downloaded to explore the differentially expressed genes (DEGs) between OA samples and RA samples. Meanwhile, metabolomic dataset MTBLS564 was downloaded and preprocessed to obtain metabolites. Then, the principal component analysis (PCA) and linear models were used to reveal DEG-metabolite relations. Finally, metabolic pathway enrichment analysis was performed to investigate the differences between the molecular mechanisms of OA and RA. Results A total of 976 DEGs and 171 metabolites were explored between OA samples and RA samples. The PCA and linear module analysis investigated 186 DEG-metabolite interactions including Glycogenin 1- (GYG1-) asparagine_54, hedgehog acyltransferase- (HHAT-) glucose_70, and TNF receptor-associated factor 3- (TRAF3-) acetoacetate_35. Finally, the KEGG pathway analysis showed that these metabolites were mainly enriched in pathways like gap junction, phagosome, NF-kappa B, and IL-17 pathway. Conclusions Genes such as HHAT, GYG1, and TRAF3, as well as metabolites including glucose, asparagine, and acetoacetate, might be implicated in the pathogenesis of OA and RA. Metabolites like ethanol and tyrosine might participate differentially in OA and RA progression via the gap junction pathway and phagosome pathway, respectively. TRAF3-acetoacetate interaction may be involved in regulating inflammation in OA and RA by the NF-kappa B and IL-17 pathway.
Collapse
|
43
|
Ciccone CD. Geriatric Pharmacology. GUCCIONE'S GERIATRIC PHYSICAL THERAPY 2020:102-136. [DOI: 10.1016/b978-0-323-60912-8.00006-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
44
|
Liu B, Chen J, Zhang S. Emerging role of ubiquitin-specific protease 14 in oncogenesis and development of tumor: Therapeutic implication. Life Sci 2019; 239:116875. [PMID: 31676235 DOI: 10.1016/j.lfs.2019.116875] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 10/25/2022]
Abstract
Ubiquitin (Ub) is a small protein that can be attached to substrate proteins to direct their degradation via the proteasome. Deubiquitinating enzymes (DUBs) reverse this process by removing ubiquitin from its substrate protein. Over the past few decades, ubiquitin-specific protease 14 (USP14), a member of the DUBs, has emerged as an important player in various types of cancers. In this article, we review and summarize biological function of USP14 in tumorigenesis and multiple signaling pathways. To determine its role in cancer, we analyzed USP14 gene expression across a panel of tumors, and discussed that it could serve as a novel bio-marker in several types of cancer. And recent contributions indicated that USP14 has been shown to act as a tumor-promoting gene via the AKT, NF-κB, MAPK pathways etc. Besides, drugs targeting USP14 have shown potential anti-tumor effect and clinical significance. We focus on recent studies that explore the link between USP14 and cancer, and further discuss USP14 as a novel target for cancer therapy.
Collapse
Affiliation(s)
- Bing Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China; State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signal Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiangping Chen
- School of International Studies, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Song Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China.
| |
Collapse
|
45
|
Mysliwiec H, Harasim-Symbor E, Baran A, Szterling-Jaworowska M, Milewska AJ, Chabowski A, Flisiak I. Abnormal serum fatty acid profile in psoriatic arthritis. Arch Med Sci 2019; 15:1407-1414. [PMID: 31749868 PMCID: PMC6855161 DOI: 10.5114/aoms.2019.89451] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/14/2017] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic inflammatory disease associated with higher risk of cardiovascular events and metabolic syndrome than psoriasis without arthritis. Fatty acids (FA) play an important role as signaling molecules in inflammatory and metabolic pathways. The aim of the study was to evaluate serum FA concentration in patients with PsA and to investigate the correlations of FA with the clinical and biochemical markers. MATERIAL AND METHODS We measured 14 FA serum concentrations by gas-liquid chromatography and flame-ionization detector after direct transesterification in 54 psoriatic patients (including 14 PsA patients) and 32 healthy controls. FA were divided according to their biologic properties into: saturated FA (SFA) and unsaturated FA (UFA), subdivided into monounsaturated FA (MUFA) and polyunsaturated FA (PUFA). RESULTS The results were correlated with Psoriasis Area and Severity Index (PASI), inflammatory and biochemical markers and lipid profile. We observed an abnormal FA profile in both psoriasis and PsA. We demonstrated lower concentrations of 10 FA in psoriasis and 7 in PsA. Patients with joint disease had a significantly higher percentage of SFA (p = 0.016) and MUFA (p = 0.001) and lower percentage of PUFA (p < 0.001) than the control group. The SFA/UFA ratio was significantly higher (p = 0.02) in PsA than in psoriasis and the controls. In the group of PsA the concentrations of docosahexaenoic acid (DHA) (p = 0.027) and n-3 PUFA (p = 0.031) correlated inversely with PASI. CONCLUSIONS Our findings indicate a changed FA profile both in psoriasis and PsA and reflect metabolic status that may predispose to the development of metabolic syndrome.
Collapse
Affiliation(s)
- Hanna Mysliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Baran
- Department of Dermatology and Venereology, Medical University of Bialystok, Bialystok, Poland
| | | | - Anna J Milewska
- Department of Statistics and Medical Informatics, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
46
|
Martínez-Zaldívar C, Azaryah H, García-Santos JA, Demmelmair H, Altmäe S, Reischl E, Rzehak P, Koletzko B, Campoy C. Early nutrition in combination with polymorphisms in fatty acid desaturase gene cluster modulate fatty acid composition of cheek cells' glycerophospholipids in school-age children. Br J Nutr 2019; 122:S68-S79. [PMID: 31638497 DOI: 10.1017/s0007114518002623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Variants in the human genes of fatty acid (FA) desaturase 1 (FADS1), 2 (FADS2) and 3 (FADS3) are associated with PUFA blood levels. We explored if maternal prenatal supplementation and children's genetic variation in seventeen SNP of the FADS1, FADS2 and FADS3 gene cluster influence twenty-one of the most relevant cheek cells' derived FA in glycerophospholipids (GPL-FA). The study was conducted in 147 Spanish and German mother-children pairs participating in the Nutraceuticals for a Healthier Life (NUHEAL) study at 8, 9 and 9·5 years. Linear and mixed model longitudinal regression analyses were performed. Maternal fish-oil (FO) or FO+5-methyltetrahydrofolate (5-MTHF) supplementation during pregnancy was associated with a significant decrease of arachidonic acid (AA) concentrations in cheek cell GPL in the offspring, from 8 to 9·5 years; furthermore, maternal FO+5-MTHF supplementation was associated with higher n-6 docosapentaenoic acid concentrations in their children at age 8 years. FADS1 rs174556 polymorphism and different FADS2 genotypes were associated with higher concentrations of linoleic and α-linolenic acids in children; moreover, some FADS2 genotypes determined lower AA concentrations in children's cheek cells. It is suggested an interaction between type of prenatal supplementation and the offspring genetic background driving GPL-FA levels at school age. Prenatal FO supplementation, and/or with 5-MTHF, seems to stimulate n-3 and n-6 FA desaturation in the offspring, increasing long-chain PUFA concentrations at school age, but depending on children's FADS1 and FADS2 genotypes. These findings suggest potential early nutrition programming of FA metabolic pathways, but interacting with children's FADS polymorphisms.
Collapse
Affiliation(s)
- Cristina Martínez-Zaldívar
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, Avda de la Investigación 11, 18016, Granada, Spain
| | - Hatim Azaryah
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, Avda de la Investigación 11, 18016, Granada, Spain
| | - José A García-Santos
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, Avda de la Investigación 11, 18016, Granada, Spain
| | - Hans Demmelmair
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich Medical Centre, Lindwurmstr, 4, D-80337 Munich, Germany
| | - Signe Altmäe
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, Avda de la Investigación 11, 18016, Granada, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, 18016, Granada, Spain
| | - Eva Reischl
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum Muenchen, D-85764 Neuherberg, Munich, Germany
| | - Peter Rzehak
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich Medical Centre, Lindwurmstr, 4, D-80337 Munich, Germany
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University of Munich Medical Centre, Lindwurmstr, 4, D-80337 Munich, Germany
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Paediatric Research, University of Granada, Avda de la Investigación 11, 18016, Granada, Spain
- Department of Paediatrics, University of Granada, Avda de la Investigación 11, 18016, Granada, Spain
- Network of Biomedical Research Centres on Epidemiology and Public Health (CIBERESP), Institute Carlos III, Madrid, Spain
| |
Collapse
|
47
|
Mustonen AM, Käkelä R, Lehenkari P, Huhtakangas J, Turunen S, Joukainen A, Kääriäinen T, Paakkonen T, Kröger H, Nieminen P. Distinct fatty acid signatures in infrapatellar fat pad and synovial fluid of patients with osteoarthritis versus rheumatoid arthritis. Arthritis Res Ther 2019; 21:124. [PMID: 31118103 PMCID: PMC6532171 DOI: 10.1186/s13075-019-1914-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/09/2019] [Indexed: 02/07/2023] Open
Abstract
Background Infrapatellar fat pad (IFP) has recently emerged as a potential source of inflammation in knee arthropathies. It has been proposed to be one source of adipocytokines, fatty acids (FA), and FA-derived lipid mediators that could contribute to the pathophysiological processes in the knee joint. Alterations in synovial fluid (SF) lipid composition have been linked to both osteoarthritis (OA) and rheumatoid arthritis (RA). The aim of the present study was to compare the FA signatures in the IFP and SF of RA and OA patients. Methods Pairs of IFP and SF samples were collected from the same knees of RA (n = 10) and OA patients (n = 10) undergoing total joint replacement surgery. Control SF samples (n = 6) were harvested during diagnostic or therapeutic arthroscopic knee surgery unrelated to RA or OA. The FA composition in the total lipids of IFP and SF was determined by gas chromatography with flame ionization and mass spectrometric detection. Results Arthropathies resulted in a significant reduction in the SF proportions of n-6 polyunsaturated FA (PUFA), more pronouncedly in OA than in RA. OA was also characterized with reduced percentages of 22:6n-3 and lower product/precursor ratios of n-3 PUFA. The proportions of total monounsaturated FA increased in both RA and OA SF. Regarding IFP, RA patients had lower proportions of 20:4n-6, total n-6 PUFA, and 22:6n-3, as well as lower product/precursor ratios of n-3 PUFA compared to OA patients. The average chain length of SF FA decreased in both diagnoses and the double bond index in OA. Conclusions The observed complex alterations in the FA signatures could have both contributed to but also limited the inflammatory processes and cartilage destruction in the RA and OA knees.
Collapse
Affiliation(s)
- Anne-Mari Mustonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland. .,Department of Environmental and Biological Sciences, Faculty of Science and Forestry, University of Eastern Finland, P.O. Box 111, FI-80101, Joensuu, Finland.
| | - Reijo Käkelä
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland.,Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute for Life Science (HiLIFE), University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
| | - Petri Lehenkari
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland.,Department of Surgery and Medical Research Center, Oulu University Hospital (OYS), P.O. Box 21, FI-90029, Oulu, Finland
| | - Johanna Huhtakangas
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland.,Rheumatology Unit and Medical Research Center, Oulu University Hospital (OYS), P.O. Box 21, FI-90029, Oulu, Finland
| | - Sanna Turunen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland.,Department of Surgery and Medical Research Center, Oulu University Hospital (OYS), P.O. Box 21, FI-90029, Oulu, Finland
| | - Antti Joukainen
- Department of Orthopaedics, Traumatology and Hand Surgery, Kuopio University Hospital (KYS), P.O. Box 100, FI-70029, Kuopio, Finland
| | - Tommi Kääriäinen
- Department of Orthopaedics, Traumatology and Hand Surgery, Kuopio University Hospital (KYS), P.O. Box 100, FI-70029, Kuopio, Finland
| | - Tommi Paakkonen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Heikki Kröger
- Department of Orthopaedics, Traumatology and Hand Surgery, Kuopio University Hospital (KYS), P.O. Box 100, FI-70029, Kuopio, Finland
| | - Petteri Nieminen
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.,Department of Environmental and Biological Sciences, Faculty of Science and Forestry, University of Eastern Finland, P.O. Box 111, FI-80101, Joensuu, Finland
| |
Collapse
|
48
|
Hopkin SJ, Lewis JW, Krautter F, Chimen M, McGettrick HM. Triggering the Resolution of Immune Mediated Inflammatory Diseases: Can Targeting Leukocyte Migration Be the Answer? Front Pharmacol 2019; 10:184. [PMID: 30881306 PMCID: PMC6407428 DOI: 10.3389/fphar.2019.00184] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
Leukocyte recruitment is a pivotal process in the regulation and resolution of an inflammatory episode. It is vital for the protective responses to microbial infection and tissue damage, but is the unwanted reaction contributing to pathology in many immune mediated inflammatory diseases (IMIDs). Indeed, it is now recognized that patients with IMIDs have defects in at least one, if not multiple, check-points regulating the entry and exit of leukocytes from the inflamed site. In this review, we will explore our understanding of the imbalance in recruitment that permits the accumulation and persistence of leukocytes in IMIDs. We will highlight old and novel pharmacological tools targeting these processes in an attempt to trigger resolution of the inflammatory response. In this context, we will focus on cytokines, chemokines, known pro-resolving lipid mediators and potential novel lipids (e.g., sphingosine-1-phosphate), along with the actions of glucocorticoids mediated by 11-beta hydroxysteroid dehydrogenase 1 and 2.
Collapse
Affiliation(s)
- Sophie J. Hopkin
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jonathan W. Lewis
- Rheumatology Research Group, Arthritis Research UK Centre of Excellence in the Pathogenesis of Rheumatoid Arthritis, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Franziska Krautter
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Myriam Chimen
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Helen M. McGettrick
- Rheumatology Research Group, Arthritis Research UK Centre of Excellence in the Pathogenesis of Rheumatoid Arthritis, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
49
|
Combination of Cell-Penetrating Peptides with Nanoparticles for Therapeutic Application: A Review. Biomolecules 2019; 9:biom9010022. [PMID: 30634689 PMCID: PMC6359287 DOI: 10.3390/biom9010022] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 02/03/2023] Open
Abstract
Cell-penetrating peptides (CPPs), also known as protein translocation domains, membrane translocating sequences or Trojan peptides, are small molecules of 6 to 30 amino acid residues capable of penetrating biological barriers and cellular membranes. Furthermore, CPP have become an alternative strategy to overcome some of the current drug limitations and combat resistant strains since CPPs are capable of delivering different therapeutic molecules against a wide range of diseases. In this review, we address the recent conjugation of CPPs with nanoparticles, which constitutes a new class of delivery vectors with high pharmaceutical potential in a variety of diseases.
Collapse
|
50
|
Watkins BA. Endocannabinoids, exercise, pain, and a path to health with aging. Mol Aspects Med 2018; 64:68-78. [DOI: 10.1016/j.mam.2018.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/22/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022]
|