1
|
Heo B, Linh VTN, Yang J, Park R, Park S, Nam M, Yoo S, Kim W, Lee M, Jung HS. AI-Assisted Plasmonic Diagnostics Platform for Osteoarthritis and Rheumatoid Arthritis With Biomarker Quantification Using Mathematical Models. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500264. [PMID: 40159800 PMCID: PMC12051836 DOI: 10.1002/smll.202500264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are major causes of functional impairment, disability, and chronic pain, leading to a substantial rise in healthcare costs. Despite differences in pathophysiology, these diseases share overlapping features that complicate diagnosis, necessitating early, more accurate, and cost-effective diagnostic tools. This study introduces an innovative plasmonic diagnostics platform for rapid and accurate label-free diagnosis of OA and RA. The sensing platform utilizes a highly dense urchin-like gold nanoarchitecture (UGN), which enhances the surface plasmonic area to significantly amplify the Raman signal. The feasibility of the developed platform for arthritis diagnosis is demonstrated by analyzing the synovial fluid (SVF) of patients. Assisted by a machine learning model, Raman signals of OA and RA groups are successfully classified with high clinical sensitivity and specificity. Metabolic biomarkers are further investigated using mathematical models of combined Pearson correlation coefficient (PCC) and non-negative matrix factorization (NMF), suggesting valuable insights for arthritis biomarker quantification. In addition, RA severity is studied using the sensing platform by classifying results from the hematology test, achieving successful stage discrimination. This platform offers a versatile, affordable, and scalable in-clinic arthritis diagnostic solution with potential applications in diagnosing and monitoring other diseases through biofluid analysis.
Collapse
Affiliation(s)
- Boyou Heo
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
| | - Vo Thi Nhat Linh
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
| | - Jun‐Yeong Yang
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
| | - Rowoon Park
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
| | - Sung‐Gyu Park
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
| | - Min‑Kyung Nam
- Department of Biomedicine & Health SciencesDepartment of Medical Life SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591Republic of Korea
| | - Seung‐Ah Yoo
- Department of Biomedicine & Health SciencesDepartment of Medical Life SciencesCollege of MedicineThe Catholic University of KoreaSeoul06591Republic of Korea
- Center for Integrative Rheumatoid Transcriptomics and DynamicsThe Catholic University of KoreaSeoul06591Republic of Korea
| | - Wan‐Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and DynamicsThe Catholic University of KoreaSeoul06591Republic of Korea
- Department of Internal MedicineThe Catholic University of KoreaSeoul06591Republic of Korea
| | - Min‐Young Lee
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
| | - Ho Sang Jung
- Advanced Bio and Healthcare Materials Research DivisionKorea Institute of Materials Science (KIMS)Changwon51508Republic of Korea
- Advanced Materials Engineering DivisionUniversity of Science and Technology (UST)Daejeon34113Republic of Korea
- School of Convergence Science and TechnologyMedical Science and EngineeringPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| |
Collapse
|
2
|
Henry ÓC, O'Neill LAJ. Metabolic Reprogramming in Stromal and Immune Cells in Rheumatoid Arthritis and Osteoarthritis: Therapeutic Possibilities. Eur J Immunol 2025; 55:e202451381. [PMID: 40170391 PMCID: PMC11962241 DOI: 10.1002/eji.202451381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/03/2025]
Abstract
Metabolic reprogramming of stromal cells, including fibroblast-like synoviocytes (FLS) and chondrocytes, as well as osteoclasts (OCs), are involved in the inflammatory and degenerative processes underlying rheumatoid arthritis (RA) and osteoarthritis (OA). In RA, FLS exhibit mTOR activation, enhanced glycolysis and reduced oxidative phosphorylation, fuelling inflammation, angiogenesis, and cartilage degradation. In OA, chondrocytes undergo metabolic rewiring, characterised by mTOR and NF-κB activation, mitochondrial dysfunction, and increased glycolysis, which promotes matrix metalloproteinase production, extracellular matrix (ECM) degradation, and angiogenesis. Macrophage-derived immunometabolites, including succinate and itaconate further modulate stromal cell function, acting as signalling molecules that modulate inflammatory and catabolic processes. Succinate promotes inflammation whilst itaconate is anti-inflammatory, suppressing inflammatory joint disease in models. Itaconate deficiency also correlates inversely with disease severity in RA in humans. Emerging evidence highlights the potential of targeting metabolic processes as promising therapeutic strategies for connective tissue disorders.
Collapse
Affiliation(s)
- Órlaith C. Henry
- Biomedical Sciences InstituteTrinity College DublinDublinIreland
| | | |
Collapse
|
3
|
Qushawy M, Alanazi MA, Hikal WM, Amirthalingam P, Abu-Gharbieh E, Almanzalawi WS, Mortagi Y, Elsherbiny N, Elsherbini AM. Optimized Nanostructured Lipid Carriers for Metformin: Enhanced Anti-Inflammatory Activity and Protection Against Type 2 Diabetes-Induced Organ Damage. Int J Nanomedicine 2025; 20:3765-3788. [PMID: 40162331 PMCID: PMC11952062 DOI: 10.2147/ijn.s506631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Background Diabetes is a chronic metabolic disease that causes serious consequences in different organs such as the heart, kidneys, pancreas, and liver. Metformin (MTF) is a common treatment for type 2 diabetes. It controls the blood glucose level by improving insulin sensitivity and glucose absorption. MTF belongs to BCS class III, which is characterized by high solubility and low permeability. Several types of nanoparticles have been developed to overcome the permeability problem of MTF. Methods In this study, we prepared nanostructured lipid carriers (NLCs) loaded with metformin (MTF) using hot melt homogenization-ultrasonication. To select the best formulation, the prepared MTF-NLCs were evaluated for entrapment efficiency % (EE%), particle size, zeta potential, polydispersity index (PDI), and in vitro drug release. The optimized formulation was selected based on the high EE%, small particle size, high absolute zeta potential, low polydispersity index, and high in vitro drug release. The optimized formulation was evaluated for surface morphology by transmission electron microscope (TEM) and for further biochemical and histological analyses in a high-fat diet-induced type 2 diabetes mellitus (T2DM) in vivo rat model; HFD was administered (44.3-kJ/kg total energy) for four weeks, followed by a single intraperitoneal injection of streptozotocin (STZ). Rats were allocated into four groups; Diabetic (DM), DM+MTF, DM+MTF-NLC, and control group. Serum and tissue samples were processed for inflammatory markers detection and histopathology. Results The prepared MTF-NLC formulation exhibited high EE% (80.65 ± 1.95% to 99.31 ± 3.25%), small particle size (247.72±5.74nm-503.23±7.26nm), high negative zeta potential (from -31.83±0.98mV to -51.6±2.64mV), PDI value less than 0.5 for all MTF-NLCs, and controlled drug release. MTF-NLC7 appeared spherical when examined by TEM. MTF and MTF-NLC groups significantly alleviated the degenerative effects of DM in both submandibular glands (SMG) and pancreas. Additionally, treatments improved kidney and liver function reduced serum inflammatory cytokines, and tissue SMG and pancreatic immunostaining of inflammatory cytokines with favorable effects of MTF-NLCs. Moreover, the MTF-NLCs showed a significant reduction of serum inflammatory cytokines, including (TNF-α and IL-1β) and pancreatic TNF-α expression, in addition to ameliorating liver and renal functions compared to MTF alone. Conclusion The preparation of MTF as NLCs improved its permeability, enhancing its anti-inflammatory activity and providing more protection against diabetes-induced organ injury.
Collapse
Affiliation(s)
- Mona Qushawy
- Department of Pharmaceutics, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, Sinai University, Alarish, North Sinai, 45511, Egypt
| | - Mansuor A Alanazi
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Wafaa M Hikal
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
- Parasitology Laboratory, Water Pollution Research Department, Environment and Climate Change Institute, National Research Centre (NRC), Giza, 12622, Egypt
| | - Palanisamy Amirthalingam
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | | | - Yasmin Mortagi
- Department of Pharmaceutics, Faculty of Pharmacy, Sinai University, Alarish, North Sinai, 45511, Egypt
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Amira M Elsherbini
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
4
|
Basu S, Ulbricht Y, Rossol M. Healthy and premature aging of monocytes and macrophages. Front Immunol 2025; 16:1506165. [PMID: 40165963 PMCID: PMC11955604 DOI: 10.3389/fimmu.2025.1506165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Aging is associated with immunosenescence, a decline in immune functions, but also with inflammaging, a chronic, low-grade inflammation, contributing to immunosenescence. Monocytes and macrophages belong to the innate immune system and aging has a profound impact on these cells, leading to functional changes and most importantly, to the secretion of pro-inflammatory cytokines and thereby contributing to inflammaging. Rheumatoid arthritis (RA) is an autoimmune disease and age is an important risk factor for developing RA. RA is associated with the early development of age-related co-morbidities like cardiovascular manifestations and osteoporosis. The immune system of RA patients shows signs of premature aging like age-inappropriate increased production of myeloid cells, accelerated telomeric erosion, and the uncontrolled production of pro-inflammatory cytokines. In this review we discuss the influence of aging on monocytes and macrophages during healthy aging and premature aging in rheumatoid arthritis.
Collapse
Affiliation(s)
- Syamantak Basu
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
| | - Ying Ulbricht
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
| | - Manuela Rossol
- Molecular Immunology, Faculty of Health Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Environment and Natural Sciences, Brandenburg University of Technology (BTU) Cottbus-Senftenberg, Senftenberg, Germany
| |
Collapse
|
5
|
Liang Y, Cheng Y, Ji J, Liu M, Wang X, Xu L, Wang W. Regulating Rheumatoid Arthritis From the Perspective of Metabolomics: A Comprehensive Review. Int J Rheum Dis 2025; 28:e70188. [PMID: 40123289 DOI: 10.1111/1756-185x.70188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Rheumatoid arthritis (RA) is a severe inflammatory autoimmune disease with metabolic changes. RA patients have abnormalities in glycolysis, amino acid metabolism, choline metabolism, and fatty acid synthesis. The differential metabolites in individuals of RA patients and animal models were explored to find the potential biomarkers for the risk prediction, diagnosis, and prognosis of RA in the perspective of metabolism. Moreover, we discussed the changes of related metabolites after treatment with anti-rheumatic drugs, Traditional Chinese Medicine (TCM) and potential metabolites for the treatment of RA to explore promising metabolites. In addition, the immunological mechanism of TCM in the treatment of RA from the perspective of metabolism was also clarified. For the perspectives of research and application of the beneficial metabolites in clinic, relevant technologies and focuses for the future studies in the field have been proposed accordingly.
Collapse
Affiliation(s)
- Yujiao Liang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingxue Cheng
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinjun Ji
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengyao Liu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinchang Wang
- National Clinical key Specialty in Rheumatology, Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Xu
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weijie Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- National Clinical key Specialty in Rheumatology, Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Huang H, Li G, He Y, Chen J, Yan J, Zhang Q, Li L, Cai X. Cellular succinate metabolism and signaling in inflammation: implications for therapeutic intervention. Front Immunol 2024; 15:1404441. [PMID: 38933270 PMCID: PMC11200920 DOI: 10.3389/fimmu.2024.1404441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Succinate, traditionally viewed as a mere intermediate of the tricarboxylic acid (TCA) cycle, has emerged as a critical mediator in inflammation. Disruptions within the TCA cycle lead to an accumulation of succinate in the mitochondrial matrix. This excess succinate subsequently diffuses into the cytosol and is released into the extracellular space. Elevated cytosolic succinate levels stabilize hypoxia-inducible factor-1α by inhibiting prolyl hydroxylases, which enhances inflammatory responses. Notably, succinate also acts extracellularly as a signaling molecule by engaging succinate receptor 1 on immune cells, thus modulating their pro-inflammatory or anti-inflammatory activities. Alterations in succinate levels have been associated with various inflammatory disorders, including rheumatoid arthritis, inflammatory bowel disease, obesity, and atherosclerosis. These associations are primarily due to exaggerated immune cell responses. Given its central role in inflammation, targeting succinate pathways offers promising therapeutic avenues for these diseases. This paper provides an extensive review of succinate's involvement in inflammatory processes and highlights potential targets for future research and therapeutic possibilities development.
Collapse
Affiliation(s)
- Hong Huang
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gejing Li
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yini He
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Chen
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jianye Yan
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qin Zhang
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liqing Li
- Department of Rheumatology of First Hospital and 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
| | - Xiong Cai
- Department of Rheumatology of First Hospital and Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
7
|
Boissais A, Blasco H, Emond P, Lefèvre A, Bigot A, Ramdani Y, Maldent NF, Mulleman D, Pillebout E, Maillot F, Audemard-Verger A. A specific diagnostic metabolome signature in adult IgA vasculitis. Metabolomics 2024; 20:61. [PMID: 38787468 PMCID: PMC11126474 DOI: 10.1007/s11306-024-02107-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 02/28/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION IgA vasculitis diagnosis relies primarily on clinical features and is confirmed by pathological findings. To date, there is no reliable noninvasive diagnostic biomarker. OBJECTIVE We aimed to explore the baseline serum metabolome of adult patients with IgA vasculitis to identify potential diagnostic biomarkers. METHODS We performed a study comparing the serum metabolome of patients with IgA vasculitis to that of patients with inflammatory condition, namely spondyloarthritis. Serum analyses were performed by high-performance liquid chromatography-mass spectrometry. RESULTS Fifty-five patients with IgA vasculitis and 77 controls with spondyloarthritis (age- and sex-matched) were included in this study. The median age of IgA vasculitis patients was 53 years. Two-thirds of patients were female (n = 32). At the time of vasculitis diagnosis, 100% of patients had skin involvement and 69% presented with glomerulonephritis (n = 38). Joint and digestive involvement were observed in 56% (n = 31) and 42% (n = 23) of patients. Four discriminative metabolites between the two groups were identified: 1-methyladenosine, L-glutamic acid, serotonin, and thymidine. The multivariate model built from the serum metabolomes of patients with IgA vasculitis and spondyloarthritis revealed an accuracy > 90%. As this model was significant according to the permutation test (p < 0.01), independent validation showed an excellent predictive value of the test set: sensitivity 98%; specificity 98%, positive predictive value 97% and negative predictive value 98%. CONCLUSION To our knowledge, this study is the first to use the metabolomic approach for diagnostic purposes in adult IgA vasculitis, highlighting a specific diagnostic metabolome signature.
Collapse
Affiliation(s)
- Alexandre Boissais
- Department of Internal Medicine and Clinical Immunology, University Hospital Center of Tours, Tours, France.
| | - Hélène Blasco
- Biochemistry and Molecular Biology Department, University Hospital Center of Tours, Tours, France
- UMR 1253, iBrain, University of Tours, 37000, InsermTours, France
| | - Patrick Emond
- UMR 1253, iBrain, University of Tours, 37000, InsermTours, France
- In Vitro Nuclear Medicine Department, University Hospital Center of Tours, Tours, France
| | - Antoine Lefèvre
- UMR 1253, iBrain, University of Tours, 37000, InsermTours, France
| | - Adrien Bigot
- Department of Internal Medicine and Clinical Immunology, University Hospital Center of Tours, Tours, France
| | - Yanis Ramdani
- Department of Internal Medicine and Clinical Immunology, University Hospital Center of Tours, Tours, France
| | - Nicole Ferreira Maldent
- Department of Internal Medicine and Clinical Immunology, University Hospital Center of Tours, Tours, France
| | - Denis Mulleman
- Center for Molecular Biophysics, UPR CNRS 4301, Tours, France
- Department of Rheumatology, University Hospital Center of Tours, Tours, France
- Nanomedicines and Nanoprobes Department, University of Tours, Tours, France
| | | | - François Maillot
- Department of Internal Medicine and Clinical Immunology, University Hospital Center of Tours, Tours, France
- UMR 1253, iBrain, University of Tours, 37000, InsermTours, France
| | - Alexandra Audemard-Verger
- Department of Internal Medicine and Clinical Immunology, University Hospital Center of Tours, Tours, France
| |
Collapse
|
8
|
Malik S, Chakraborty D, Agnihotri P, Kumar V, Biswas S. Unveiling the Nexus: Cellular Metabolomics Unravels the Impact of Estrogen on Nicotinamide Metabolism in Mitigating Rheumatoid Arthritis Pathogenesis. Metabolites 2024; 14:214. [PMID: 38668342 PMCID: PMC11052502 DOI: 10.3390/metabo14040214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is a metabolic joint disorder influenced by hormonal regulation, notably estrogen, which plays a cytoprotective role against inflammation. While estrogen's impact on RA pathogenesis has been studied, the altered metabolite expression under estrogen's influence remains unexplored. This study investigated the changes in the metabolome of synovial fibroblasts isolated from RA patients under 17β-estradiol (E2) using the liquid chromatography with tandem mass spectrometry (LC-MS/MS) approach followed by multivariate and biological pathway analysis along with in vitro validation. Results identified 3624 m/z, among which eight metabolites were significant (p < 0.05). Nicotinate and nicotinamide metabolism was found to be highly correlated with the treatment of E2, with metabolites NAD+ and 1-methynicotinamide (1-MNA) upregulated by E2 induction in RA-FLS. PharmMapper analysis identified potential gene targets of 1-MNA, which were further matched with RA gene targets, and thus, STAT1, MAPK14, MMP3, and MMP9 were concluded to be the common targets. E2 treatment affected the expression of these gene targets and ameliorated the development of oxidative stress associated with RA inflammation, which can be attributed to increased concentration of 1-MNA. Thus, an LC-MS/MS-based metabolomics study revealed the prominent role of estrogen in preventing inflammatory progression in RA by altering metabolite concentration, which can support its therapeutic capacity in remitting RA.
Collapse
Affiliation(s)
- Swati Malik
- Department of Integrative and Functional Biology, CSIR—Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (S.M.); (D.C.); (P.A.)
- AcSIR—Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Debolina Chakraborty
- Department of Integrative and Functional Biology, CSIR—Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (S.M.); (D.C.); (P.A.)
- AcSIR—Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Prachi Agnihotri
- Department of Integrative and Functional Biology, CSIR—Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (S.M.); (D.C.); (P.A.)
- AcSIR—Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| | - Vijay Kumar
- Department of Orthopaedics, AIIMS—All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India;
| | - Sagarika Biswas
- Department of Integrative and Functional Biology, CSIR—Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; (S.M.); (D.C.); (P.A.)
- AcSIR—Academy of Scientific and Innovative Research, Ghaziabad 201002, India
| |
Collapse
|
9
|
Kim SW, Kim CW, Moon YA, Kim HS. Reprogramming of tumor-associated macrophages by metabolites generated from tumor microenvironment. Anim Cells Syst (Seoul) 2024; 28:123-136. [PMID: 38577621 PMCID: PMC10993762 DOI: 10.1080/19768354.2024.2336249] [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: 01/25/2024] [Accepted: 03/17/2024] [Indexed: 04/06/2024] Open
Abstract
The tumor microenvironment comprises both tumor and non-tumor stromal cells, including tumor-associated macrophages (TAMs), endothelial cells, and carcinoma-associated fibroblasts. TAMs, major components of non-tumor stromal cells, play a crucial role in creating an immunosuppressive environment by releasing cytokines, chemokines, growth factors, and immune checkpoint proteins that inhibit T cell activity. During tumors develop, cancer cells release various mediators, including chemokines and metabolites, that recruit monocytes to infiltrate tumor tissues and subsequently induce an M2-like phenotype and tumor-promoting properties. Metabolites are often overlooked as metabolic waste or detoxification products but may contribute to TAM polarization. Furthermore, macrophages display a high degree of plasticity among immune cells in the tumor microenvironment, enabling them to either inhibit or facilitate cancer progression. Therefore, TAM-targeting has emerged as a promising strategy in tumor immunotherapy. This review provides an overview of multiple representative metabolites involved in TAM phenotypes, focusing on their role in pro-tumoral polarization of M2.
Collapse
Affiliation(s)
- Seung Woo Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Chan Woo Kim
- Cancer Immunotherapy Evaluation Team, Non-Clinical Evaluation Center, Osong Medical Innovation Foundation (KBIO Health), Cheongju, Republic of Korea
| | - Young-Ah Moon
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
10
|
Tian Q, Ruan J, Wang Y, Xiao Y, Cheng Q, Chen Y, Li M, Chang K, Yi X. Extracellular succinate derived from ectopic milieu drives adhesion and implantation growth of ectopic endometrial stromal cells via the SUCNR1 signal in endometriosis. Cell Commun Signal 2024; 22:82. [PMID: 38291428 PMCID: PMC10826047 DOI: 10.1186/s12964-023-01415-7] [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: 08/28/2023] [Accepted: 12/02/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND As a dual-function metabolite, succinate has emerged in cell function and plays a key signaling role in linking mitochondrial function to other cellular functions. Succinate accumulation in the cytoplasm is commonly associated with hypoxia in the microenvironment and immune cell activation. Extracellular succinate released into the microenvironment is considered an inflammatory alarm that can be sensed by its membrane receptor SUCNR1, which boosts proinflammatory responses and acts akin to classical hormones and cytokines. Succinate plays an important role in the development of inflammatory diseases. Whether succinate facilitates the progression of endometriosis (EMs), characterized by chronic inflammation and peritoneal adhesion, is worth exploring. OBJECTIVE We mimicked the ectopic milieu in vitro and in vivo to evaluate the main source and potential role of succinate in endometriosis. We assessed the molecular and functional effects of succinate on macrophages and peritoneal mesothelial cells in peritoneal cavity. The effect of succinate/SUCNR1 signaling on ectopic endometrial stromal cells (ESCs) was further explored in this study. METHODS In this study, we used targeted organic acid metabolomics analysis and in vitro assays to assess the potential accumulation of succinate in the peritoneal fluid of EMs patients. We examined its correlation with disease severity, Visual Analogue Scale, and the Endometriosis Fertility Index. Flow cytometry, enzyme linked immunosorbent assay, western blot assay, quantitative real-time PCR, and other molecular biology techniques were used to explore the potential mechanisms. RESULTS By mimicking the ectopic milieu, we constructed an in vitro co-culture system and found that M1 polarized macrophages and that the peritoneal mesothelial cell line (HMrSV5) mainly released succinate into their microenvironment and activated the succinate receptor (SUCNR1) signal, which further polarized the macrophages and significantly enhanced the invasive survival of ESCs, and the adhesion to the peritoneum. We further investigated the pathological effects of extracellular succinate in vivo using a xenograft mouse models of endometriosis. CONCLUSIONS Succinate-SUCNR1 signaling facilitates the creation of inflammatory cells and plays a vital role in EMs progression and peritoneal adhesion. Our work on the molecular mechanisms underlying succinate accumulation and function will help elucidate the phenotypic mysteries of pain and infertility in EMs. Video Abstract.
Collapse
Affiliation(s)
- Qi Tian
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Jingyao Ruan
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yuning Wang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yinping Xiao
- Department of Pathology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Qi Cheng
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yun Chen
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Kaikai Chang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| | - Xiaofang Yi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| |
Collapse
|
11
|
Meng Q, Lin M, Song W, Wu J, Cao G, Huang P, Su Z, Gu W, Deng X, Xu P, Yang Y, Li H, Liu H, Zhang F. The gut-joint axis mediates the TNF-induced RA process and PBMT therapeutic effects through the metabolites of gut microbiota. Gut Microbes 2023; 15:2281382. [PMID: 38017660 PMCID: PMC10730145 DOI: 10.1080/19490976.2023.2281382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
The gut-joint axis, one of the mechanisms that mediates the onset and progression of joint and related diseases through gut microbiota, and shows the potential as therapeutic target. A variety of drugs exert therapeutic effects on rheumatoid arthritis (RA) through the gut-joint axis. However, the anti-inflammatory and immunomodulatory effect of novel photobiomodulatory therapy (PBMT) on RA need further validation and the involvement of gut-joint axis in this process remains unknown. The present study demonstrated the beneficial effects of PBMT on RA, where we found the restoration of gut microbiota homeostasis, and the related key pathways and metabolites after PBMT. We also discovered that the therapeutic effects of PBMT on RA mainly through the gut-joint axis, in which the amino acid metabolites (Alanine and N-acetyl aspartate) play the key role and rely on the activity of metabolic enzymes in the target organs. Together, the results prove that the metabolites of amino acid from gut microbiota mediate the regulation effect on the gut-joint axis and the therapeutic effect on rheumatoid arthritis of PBMT.
Collapse
Affiliation(s)
- Qingtai Meng
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Monan Lin
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Wuqi Song
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Jiahui Wu
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Guoding Cao
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Ping Huang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Zaiyu Su
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Wei Gu
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Xueqing Deng
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Peng Xu
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Yi Yang
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Hui Li
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Hailiang Liu
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Fengmin Zhang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Zheng X, Zhu Y, Zhao Z, Chu Y, Yang W. The role of amino acid metabolism in inflammatory bowel disease and other inflammatory diseases. Front Immunol 2023; 14:1284133. [PMID: 37936710 PMCID: PMC10626463 DOI: 10.3389/fimmu.2023.1284133] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Inflammation is a characteristic symptom of the occurrence and development of many diseases, which is mainly characterized by the infiltration of inflammatory cells such as macrophages and granulocytes, and the increased release of proinflammatory factors. Subsequently, macrophage differentiates and T cells and other regulated factors exhibit anti-inflammatory function, releasing pro- and anti-inflammatory factors to maintain homeostasis. Although reports define various degrees of metabolic disorders in both the inflamed and non-inflamed parts of inflammatory diseases, little is known about the changes in amino acid metabolism in such conditions. This review aims to summarize amino acid changes and mechanisms involved in the progression of inflammatory bowel disease (IBD) and other inflammatory diseases. Since mesenchymal stem cells (MSCs) and their derived exosomes (MSC-EXO) have been found to show promising effects in the treatment of IBD and other inflammatory diseases,their potential in the modulation of amino acid metabolism in the treatment of inflammation is also discussed.
Collapse
Affiliation(s)
- Xiaowen Zheng
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yi Zhu
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Zihan Zhao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Chu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Wenjing Yang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
13
|
Zhang J, Sun M, Elmaidomy AH, Youssif KA, Zaki AMM, Hassan Kamal H, Sayed AM, Abdelmohsen UR. Emerging trends and applications of metabolomics in food science and nutrition. Food Funct 2023; 14:9050-9082. [PMID: 37740352 DOI: 10.1039/d3fo01770b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
The study of all chemical processes involving metabolites is known as metabolomics. It has been developed into an essential tool in several disciplines, such as the study of plant physiology, drug development, human diseases, and nutrition. The field of food science, diagnostic biomarker research, etiological analysis in the field of medical therapy, and raw material quality, processing, and safety have all benefited from the use of metabolomics recently. Food metabolomics includes the use of metabolomics in food production, processing, and human diets. As a result of changing consumer habits and the rising of food industries all over the world, there is a remarkable increase in interest in food quality and safety. It requires the employment of various technologies for the food supply chain, processing of food, and even plant breeding. This can be achieved by understanding the metabolome of food, including its biochemistry and composition. Additionally, Food metabolomics can be used to determine the similarities and differences across crop kinds, as an indicator for tracking the process of ripening to increase crops' shelf life and attractiveness, and identifying metabolites linked to pathways responsible for postharvest disorders. Moreover, nutritional metabolomics is used to investigate the connection between diet and human health through detection of certain biomarkers. This review assessed and compiled literature on food metabolomics research with an emphasis on metabolite extraction, detection, and data processing as well as its applications to the study of food nutrition, food-based illness, and phytochemical analysis. Several studies have been published on the applications of metabolomics in food but further research concerning the use of standard reproducible procedures must be done. The results published showed promising uses in the food industry in many areas such as food production, processing, and human diets. Finally, metabolome-wide association studies (MWASs) could also be a useful predictor to detect the connection between certain diseases and low molecular weight biomarkers.
Collapse
Affiliation(s)
- Jianye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Mingna Sun
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Abeer H Elmaidomy
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Khayrya A Youssif
- Department of Pharmacognosy, Faculty of Pharmacy, El-Saleheya El Gadida University, Cairo, Egypt
| | - Adham M M Zaki
- Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Hossam Hassan Kamal
- Faculty of Pharmacy, Deraya University, 7 Universities Zone, New Minia 61111, Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, 62513 Beni-Suef, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Almaaqal University, 61014 Basra, Iraq
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, 7 Universities Zone, New Minia 61111, Egypt
| |
Collapse
|
14
|
Jeljeli MM, Adamopoulos IE. Innate immune memory in inflammatory arthritis. Nat Rev Rheumatol 2023; 19:627-639. [PMID: 37674048 PMCID: PMC10721491 DOI: 10.1038/s41584-023-01009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 09/08/2023]
Abstract
The concept of immunological memory was demonstrated in antiquity when protection against re-exposure to pathogens was observed during the plague of Athens. Immunological memory has been linked with the adaptive features of T and B cells; however, in the past decade, evidence has demonstrated that innate immune cells can exhibit memory, a phenomenon called 'innate immune memory' or 'trained immunity'. Innate immune memory is currently being defined and is transforming our understanding of chronic inflammation and autoimmunity. In this Review, we provide an up-to-date overview of the memory-like features of innate immune cells in inflammatory arthritis and the crosstalk between chronic inflammatory milieu and cell reprogramming. Aberrant pro-inflammatory signalling, including cytokines, regulates the metabolic and epigenetic reprogramming of haematopoietic progenitors, leading to exacerbated inflammatory responses and osteoclast differentiation, in turn leading to bone destruction. Moreover, imprinted memory on mature cells including terminally differentiated osteoclasts alters responsiveness to therapies and modifies disease outcomes, commonly manifested by persistent inflammatory flares and relapse following medication withdrawal.
Collapse
Affiliation(s)
- Maxime M Jeljeli
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Iannis E Adamopoulos
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
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: 31] [Impact Index Per Article: 15.5] [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
|
16
|
Suwa Y, Nagafuchi Y, Yamada S, Fujio K. The role of dendritic cells and their immunometabolism in rheumatoid arthritis. Front Immunol 2023; 14:1161148. [PMID: 37251399 PMCID: PMC10213288 DOI: 10.3389/fimmu.2023.1161148] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Dendritic cells (DCs) play crucial roles in the pathogenesis of rheumatoid arthritis (RA), a prototypic autoimmune disease characterized by chronic synovitis and joint destruction. Conventional dendritic cells (cDCs) with professional antigen-presenting functions are enriched in the RA synovium. In the synovium, the cDCs are activated and show both enhanced migratory capacities and T cell activation in comparison with peripheral blood cDCs. Plasmacytoid dendritic cells, another subtype of DCs capable of type I interferon production, are likely to be tolerogenic in RA. Monocyte-derived dendritic cells (moDCs), once called "inflammatory DCs", are localized in the RA synovium, and they induce T-helper 17 cell expansion and enhanced proinflammatory cytokine production. Recent studies revealed that synovial proinflammatory hypoxic environments are linked to metabolic reprogramming. Activation of cDCs in the RA synovium is accompanied by enhanced glycolysis and anabolism. In sharp contrast, promoting catabolism can induce tolerogenic DCs from monocytes. Herein, we review recent studies that address the roles of DCs and their immunometabolic features in RA. Immunometabolism of DCs could be a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Yuichi Suwa
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Saeko Yamada
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Mustonen AM, Tollis S, Käkelä R, Sihvo SP, Palosaari S, Pohjanen VM, Yli-Hallila A, Lehenkari P, Nieminen P. Increased n-6 Polyunsaturated Fatty Acids Indicate Pro- and Anti-Inflammatory Lipid Modifications in Synovial Membranes with Rheumatoid Arthritis. Inflammation 2023:10.1007/s10753-023-01816-3. [PMID: 37140681 PMCID: PMC10359413 DOI: 10.1007/s10753-023-01816-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/05/2023]
Abstract
Emerging evidence suggests that fatty acids (FAs) and their lipid mediator derivatives can induce both beneficial and detrimental effects on inflammatory processes and joint degradation in osteoarthritis (OA) and autoimmune-driven rheumatoid arthritis (RA). The present study characterized the detailed FA signatures of synovial membranes collected during knee replacement surgery of age- and gender-matched OA and RA patients (n = 8/diagnosis). The FA composition of total lipids was determined by gas chromatography and analyzed with univariate and multivariate methods supplemented with hierarchical clustering (HC), random forest (RF)-based classification of FA signatures, and FA metabolism pathway analysis. RA synovium lipids were characterized by reduced proportions of shorter-chain saturated FAs (SFAs) and elevated percentages of longer-chain SFAs and monounsaturated FAs, alkenyl chains, and C20 n-6 polyunsaturated FAs compared to OA synovium lipids. In HC, FAs and FA-derived variables clustered into distinct groups, which preserved the discriminatory power of the individual variables in predicting the RA and OA inflammatory states. In RF classification, SFAs and 20:3n-6 were among the most important FAs distinguishing RA and OA. Pathway analysis suggested that elongation reactions of particular long-chain FAs would have increased relevance in RA. The present study was able to determine the individual FAs, FA groups, and pathways that distinguished the more inflammatory RA from OA. The findings suggest modifications of FA elongation and metabolism of 20:4n-6, glycerophospholipids, sphingolipids, and plasmalogens in the chronically inflamed RA synovium. These FA alterations could have implications in lipid mediator synthesis and potential as novel diagnostic and therapeutic tools.
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, Forestry and Technology, University of Eastern Finland, P.O. Box 111, FI-80101, Joensuu, Finland.
| | - Sylvain Tollis
- Institute of Biomedicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, 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 of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
| | - Sanna P Sihvo
- 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 of Life Science (HiLIFE) and Biocenter Finland, University of Helsinki, P.O. Box 65, FI-00014, Helsinki, Finland
| | - Sanna Palosaari
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
- Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014, Oulu, Finland
| | - Vesa-Matti Pohjanen
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
- Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014, Oulu, Finland
| | - Aaron Yli-Hallila
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
| | - Petri Lehenkari
- Cancer and Translational Medicine Research Unit, Faculty of Medicine, University of Oulu, P.O. Box 5000, FI-90014, Oulu, Finland
- Medical Research Center, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014, Oulu, Finland
- Department of Surgery, Oulu University Hospital, P.O. Box 21, FI-90029, OYS, Oulu, 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
| |
Collapse
|
18
|
Cedeno M, Murillo-Saich J, Coras R, Cedola F, Brandy A, Prior A, Pedersen A, Mateo L, Martinez-Morillo M, Guma M. Serum metabolomic profiling identifies potential biomarkers in arthritis in older adults: an exploratory study. Metabolomics 2023; 19:37. [PMID: 37022535 PMCID: PMC11449491 DOI: 10.1007/s11306-023-02004-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Seronegative elderly-onset rheumatoid arthritis (EORA)neg and polymyalgia rheumatica (PMR) have similar clinical characteristics making them difficult to distinguish based on clinical features. We hypothesized that the study of serum metabolome could identify potential biomarkers of PMR vs. EORAneg. METHODS Arthritis in older adults (ARTIEL) is an observational prospective cohort with patients older than 60 years of age with newly diagnosed arthritis. Patients' blood samples were compared at baseline with 18 controls. A thorough clinical examination was conducted. A Bruker Avance 600 MHz spectrometer was used to acquire Nuclear Magnetic Resonance (NMR) spectra of serum samples. Chenomx NMR suite 8.5 was used for metabolite identification and quantification.Student t-test, one-way ANOVA, binary linear regression and ROC curve, Pearson's correlation along with pathway analyses were conducted. RESULTS Twenty-eight patients were diagnosed with EORAneg and 20 with PMR. EORAneg patients had a mean disease activity score (DAS)-Erythrocyte Sedimentation Rate (ESR) of 6.21 ± 1.00. All PMR patients reported shoulder pain, and 90% reported pelvic pain. Fifty-eight polar metabolites were identified. Of these, 3-hydroxybutyrate, acetate, glucose, glycine, lactate, and o-acetylcholine (o-ACh), were significantly different between groups. Of interest, IL-6 correlated with different metabolites in PMR and EORAneg suggesting different inflammatory activated pathways. Finally, lactate, o-ACh, taurine, and sex (female) were identified as distinguishable factors of PMR from EORAneg with a sensitivity of 90%, specificity of 92.3%, and an AUC of 0.925 (p < 0.001). CONCLUSION These results suggest that EORAneg and PMR have different serum metabolomic profiles that might be related to their pathobiology and can be used as biomarker to discriminate between both diseases.
Collapse
Affiliation(s)
- Martha Cedeno
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jessica Murillo-Saich
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Roxana Coras
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, Bellaterra, Barcelona, 08193, Spain
| | - Francesca Cedola
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Anahy Brandy
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain
| | - Agueda Prior
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain
| | - Anders Pedersen
- Swedish NMR Centre, University of Gothenburg, Gothenburg, 41390, Sweden
| | - Lourdes Mateo
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain
| | - Melania Martinez-Morillo
- Department of Rheumatology, Germans Trias i Pujol, University Hospital, Carretera de Canyet, Badalona, 08916, Spain.
| | - Monica Guma
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, Bellaterra, Barcelona, 08193, Spain.
- VA Healthcare Service, San Diego, CA, 92161, USA.
| |
Collapse
|
19
|
Innate and adaptive immune abnormalities underlying autoimmune diseases: the genetic connections. SCIENCE CHINA. LIFE SCIENCES 2023:10.1007/s11427-021-2187-3. [PMID: 36738430 PMCID: PMC9898710 DOI: 10.1007/s11427-021-2187-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023]
Abstract
With the exception of an extremely small number of cases caused by single gene mutations, most autoimmune diseases result from the complex interplay between environmental and genetic factors. In a nutshell, etiology of the common autoimmune disorders is unknown in spite of progress elucidating certain effector cells and molecules responsible for pathologies associated with inflammatory and tissue damage. In recent years, population genetics approaches have greatly enriched our knowledge regarding genetic susceptibility of autoimmunity, providing us with a window of opportunities to comprehensively re-examine autoimmunity-associated genes and possible pathways. In this review, we aim to discuss etiology and pathogenesis of common autoimmune disorders from the perspective of human genetics. An overview of the genetic basis of autoimmunity is followed by 3 chapters detailing susceptibility genes involved in innate immunity, adaptive immunity and inflammatory cell death processes respectively. With such attempts, we hope to expand the scope of thinking and bring attention to lesser appreciated molecules and pathways as important contributors of autoimmunity beyond the 'usual suspects' of a limited subset of validated therapeutic targets.
Collapse
|
20
|
Zhang Z, Zhang J, Zhao S, Peng T, Zhang M, Xu X, Li K, Liu Y. Synovial tissue metabolomics analysis of the therapeutic effects of stir-fried Xanthii Fructus on rheumatoid arthritis in rats. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1216:123594. [PMID: 36638684 DOI: 10.1016/j.jchromb.2023.123594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/23/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Rheumatoid arthritis (RA) is a rheumatic disease that easily causes synovial hyperplasia and joint damage. Comprehensive metabolomic profiling of synovial tissue can reveal local pathological changes during RA and identify metabolites as candidate biomarkers. Detecting metabolites in synovial tissue can more directly reflect the pathological state and disease activity associated with it. stir-fried Xanthii Fructus has demonstrated efficacy in treating RA, but its pharmacodynamic property and mechanism of action are unclear. In this study, the molecular composition of the extract of stir-fried Xanthium Fructus was determined through HPLC. The major components that exert anti-inflammatory and analgesic effects were speculated to be phenolic acids. Next, the effect of stir-fried Xanthii Fructus extracts in RA treatment was comprehensively evaluated using rat body weight, foot volume, inflammatory factors, and histopathological sections of the ankle joint as evaluation indicators. The results showed that the extract of stir-fried Xanthii Fructus could significantly reduce the inflammatory response and improve the degree of joint swelling and the imbalance between pro-inflammatory and anti-inflammatory in adjuvant arthritis rats. Finally, non-targeted metabolomics based on UPLC-Q-TOF/MS and multivariate statistical analysis were used to explore the changes of endogenous metabolites in synovium tissues and to search for potential biomarkers and related metabolic pathways in stir-fried Xanthii Fructus extract-treated AA rats. The results showed that stir-fried Xanthii Fructus mainly treated RA by regulating energy metabolism, hormone metabolism, amino acid metabolism and oxidative stress response in adjuvant arthritis rats. This study provides a theoretical basis for the mechanism of action of stir-fried Xanthii Fructus extract in treating RA.
Collapse
Affiliation(s)
- Zuohua Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China; Infection Control Section, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Jinpeng Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China
| | - Shuo Zhao
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China
| | - Tingtao Peng
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China
| | - Mei Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China
| | - Xizhu Xu
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China
| | - Ke Li
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China.
| | - Yuanyuan Liu
- Department of Pharmaceutical Analysis, College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271099, China.
| |
Collapse
|
21
|
Abstract
Succinate is a circulating metabolite, and the relationship between abnormal changes in the physiological concentration of succinate and inflammatory diseases caused by the overreaction of certain immune cells has become a research focus. Recent investigations have shown that succinate produced by the gut microbiota has the potential to regulate host homeostasis and treat diseases such as inflammation. Gut microbes are important for maintaining intestinal homeostasis. Microbial metabolites serve as nutrients in energy metabolism, and act as signal molecules that stimulate host cell and organ function and affect the structural balance between symbiotic gut microorganisms. This review focuses on succinate as a metabolite of both host cells and gut microbes and its involvement in regulating the gut - immune tissue axis by activating intestinal mucosal cells, including macrophages, dendritic cells, and intestinal epithelial cells. We also examined its role as the mediator of microbiota - host crosstalk and its potential function in regulating intestinal microbiota homeostasis. This review explores feasible ways to moderate succinate levels and provides new insights into succinate as a potential target for microbial therapeutics for humans.
Collapse
Affiliation(s)
- Yi-Han Wei
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiang-Chao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| |
Collapse
|
22
|
Acid sensor ASIC1a induces synovial fibroblast proliferation via Wnt/β-catenin/c-Myc pathway in rheumatoid arthritis. Int Immunopharmacol 2022; 113:109328. [DOI: 10.1016/j.intimp.2022.109328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/25/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
23
|
Thibaut R, Orliaguet L, Ejlalmanesh T, Venteclef N, Alzaid F. Perspective on direction of control: Cellular metabolism and macrophagepolarization. Front Immunol 2022; 13:918747. [PMID: 36159824 PMCID: PMC9493491 DOI: 10.3389/fimmu.2022.918747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages are innate immune cells with high phenotypic plasticity. Depending on the microenvironmental cues they receive, they polarize on a spectrum with extremes being pro- or anti-inflammatory. As well as responses to microenvironmental cues, cellular metabolism is increasingly recognized as a key factor influencing macrophage function. While pro-inflammatory macrophages mostly use glycolysis to meet their energetic needs, anti-inflammatory macrophages heavily rely on mitochondrial respiration. The relationship between macrophage phenotype and macrophage metabolism is well established, however its precise directionality is still under question. Indeed, whether cellular metabolism per se influences macrophage phenotype or whether macrophage polarization dictates metabolic activity is an area of active research. In this short perspective article, we sought to shed light on this area. By modulating several metabolic pathways in bone marrow-derived macrophages, we show that disruption of cellular metabolism does per se influence cytokine secretion profile and expression of key inflammatory genes. Only some pathways seem to be involved in these processes, highlighting the need for specific metabolic functions in the regulation of macrophage phenotype. We thus demonstrate that the intact nature of cellular metabolism influences macrophage phenotype and function, addressing the directionality between these two aspects of macrophage biology.
Collapse
Affiliation(s)
- Ronan Thibaut
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | - Lucie Orliaguet
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | - Tina Ejlalmanesh
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | - Nicolas Venteclef
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
| | - Fawaz Alzaid
- INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Institut Necker Enfants Malades, Paris, France
- Dasman Diabetes Institute, Kuwait, Kuwait
- *Correspondence: Fawaz Alzaid,
| |
Collapse
|
24
|
Srimadh Bhagavatham SK, Pulukool SK, Pradhan SS, R S, Ashok Naik A, V M DD, Sivaramakrishnan V. Systems biology approach delineates critical pathways associated with disease progression in rheumatoid arthritis. J Biomol Struct Dyn 2022:1-22. [PMID: 36047508 DOI: 10.1080/07391102.2022.2115555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Rheumatoid Arthritis (RA) is a chronic systemic autoimmune disease leading to inflammation, cartilage cell death, synoviocyte proliferation, and increased and impaired differentiation of osteoclasts and osteoblasts leading to joint erosions and deformities. Transcriptomics, proteomics, and metabolomics datasets were analyzed to identify the critical pathways that drive the RA pathophysiology. Single nucleotide polymorphisms (SNPs) associated with RA were analyzed for the functional implications, clinical outcomes, and blood parameters later validated by literature. SNPs associated with RA were grouped into pathways that drive the immune response and cytokine production. Further gene set enrichment analysis (GSEA) was performed on gene expression omnibus (GEO) data sets of peripheral blood mononuclear cells (PBMCs), synovial macrophages, and synovial biopsies from RA patients showed enrichment of Th1, Th2, Th17 differentiation, viral and bacterial infections, metabolic signalling and immunological pathways with potential implications for RA. The proteomics data analysis presented pathways with genes involved in immunological signaling and metabolic pathways, including vitamin B12 and folate metabolism. Metabolomics datasets analysis showed significant pathways like amino-acyl tRNA biosynthesis, metabolism of amino acids (arginine, alanine aspartate, glutamate, glutamine, phenylalanine, and tryptophan), and nucleotide metabolism. Furthermore, our commonality analysis of multi-omics datasets identified common pathways with potential implications for joint remodeling in RA. Disease-modifying anti-rheumatic drugs (DMARDs) and biologics treatments were found to modulate many of the pathways that were deregulated in RA. Overall, our analysis identified molecular signatures associated with the observed symptoms, joint erosions, potential biomarkers, and therapeutic targets in RA. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Sujith Kumar Pulukool
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Saiswaroop R
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Ashwin Ashok Naik
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Datta Darshan V M
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur, A.P., India
| |
Collapse
|
25
|
Xu L, Chang C, Jiang P, Wei K, Zhang R, Jin Y, Zhao J, Xu L, Shi Y, Guo S, He D. Metabolomics in rheumatoid arthritis: Advances and review. Front Immunol 2022; 13:961708. [PMID: 36032122 PMCID: PMC9404373 DOI: 10.3389/fimmu.2022.961708] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/25/2022] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease accompanied by metabolic alterations. The metabolic profiles of patients with RA can be determined using targeted and non-targeted metabolomics technology. Metabolic changes in glucose, lipid, and amino acid levels are involved in glycolysis, the tricarboxylic acid cycle, the pentose phosphate pathway, the arachidonic acid metabolic pathway, and amino acid metabolism. These alterations in metabolic pathways and metabolites can fulfill bio-energetic requirements, promote cell proliferation, drive inflammatory mediator secretion, mediate leukocyte infiltration, induce joint destruction and muscle atrophy, and regulate cell proliferation, which may reflect the etiologies of RA. Differential metabolites can be used as biomarkers for the diagnosis, prognosis, and risk prediction, improving the specificity and accuracy of diagnostics and prognosis prediction. Additionally, metabolic changes associated with therapeutic responses can improve the understanding of drug mechanism. Metabolic homeostasis and regulation are new therapeutic strategies for RA. In this review, we provide a comprehensive overview of advances in metabolomics for RA.
Collapse
Affiliation(s)
- Lingxia Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Runrun Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yehua Jin
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianan Zhao
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Arra M, Swarnkar G, Adapala NS, Naqvi SK, Cai L, Rai MF, Singamaneni S, Mbalaviele G, Brophy R, Abu-Amer Y. Glutamine metabolism modulates chondrocyte inflammatory response. eLife 2022; 11:e80725. [PMID: 35916374 PMCID: PMC9371604 DOI: 10.7554/elife.80725] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Osteoarthritis is the most common joint disease in the world with significant societal consequences but lacks effective disease-modifying interventions. The pathophysiology consists of a prominent inflammatory component that can be targeted to prevent cartilage degradation and structural defects. Intracellular metabolism has emerged as a culprit of the inflammatory response in chondrocytes, with both processes co-regulating each other. The role of glutamine metabolism in chondrocytes, especially in the context of inflammation, lacks a thorough understanding and is the focus of this work. We display that mouse chondrocytes utilize glutamine for energy production and anabolic processes. Furthermore, we show that glutamine deprivation itself causes metabolic reprogramming and decreases the inflammatory response of chondrocytes through inhibition of NF-κB activity. Finally, we display that glutamine deprivation promotes autophagy and that ammonia is an inhibitor of autophagy. Overall, we identify a relationship between glutamine metabolism and inflammatory signaling and display the need for increased study of chondrocyte metabolic systems.
Collapse
Affiliation(s)
- Manoj Arra
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Gaurav Swarnkar
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Naga Suresh Adapala
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Syeda Kanwal Naqvi
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Lei Cai
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Muhammad Farooq Rai
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Material Sciences, Washington University School of Medicine, St Louis, United States
| | - Gabriel Mbalaviele
- Bone and Mineral Division, Department of Medicine, Washington University School of Medicine, St Louis, United States
| | - Robert Brophy
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
| | - Yousef Abu-Amer
- Department of Orthopedic Surgery, Washington University School of Medicine, St Louis, United States
- Shriners Hospital for Children, Saint Louis, United States
| |
Collapse
|
27
|
Kim S, Hwang J, Kim J, Lee SH, Cheong YE, Lee S, Kim KH, Cha HS. Metabolic discrimination of synovial fluid between rheumatoid arthritis and osteoarthritis using gas chromatography/time-of-flight mass spectrometry. Metabolomics 2022; 18:48. [PMID: 35781849 DOI: 10.1007/s11306-022-01893-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/26/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) and osteoarthritis (OA) are clinicopathologically different. OBJECTIVES We aimed to assess the feasibility of metabolomics in differentiating the metabolite profiles of synovial fluid between RA and OA using gas chromatography/time-of-flight mass spectrometry. METHODS We first compared the global metabolomic changes in the synovial fluid of 19 patients with RA and OA. Partial least squares-discriminant, hierarchical clustering, and univariate analyses were performed to distinguish metabolites of RA and OA. These findings were then validated using synovial fluid samples from another set of 15 patients with RA and OA. RESULTS We identified 121 metabolites in the synovial fluid of the first 19 samples. The score plot of PLS-DA showed a clear separation between RA and OA. Twenty-eight crucial metabolites, including hypoxanthine, xanthine, adenosine, citrulline, histidine, and tryptophan, were identified to be capable of distinguishing RA metabolism from that of OA; these were found to be associated with purine and amino acid metabolism. CONCLUSION Our results demonstrated that metabolite profiling of synovial fluid could clearly discriminate between RA and OA, suggesting that metabolomics may be a feasible tool to assist in the diagnosis and advance the comprehension of pathological processes for diseases.
Collapse
Affiliation(s)
- Sooah Kim
- Department of Environment Science & Biotechnology, Jeonju University, Jeonju, 55069, Republic of Korea
| | - Jiwon Hwang
- Division of Rheumatology, Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, 51353, Republic of Korea
| | - Jungyeon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Sun-Hee Lee
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Yu Eun Cheong
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Seulkee Lee
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Hoon-Suk Cha
- Division of Rheumatology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea.
| |
Collapse
|
28
|
Welhaven HD, Vahidi G, Walk ST, Bothner B, Martin SA, Heveran CM, June RK. The cortical bone metabolome of
C57BL
/
6J
mice is sexually dimorphic. JBMR Plus 2022; 6:e10654. [PMID: 35866150 PMCID: PMC9289981 DOI: 10.1002/jbm4.10654] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cortical bone quality, which is sexually dimorphic, depends on bone turnover and therefore on the activities of remodeling bone cells. However, sex differences in cortical bone metabolism are not yet defined. Adding to the uncertainty about cortical bone metabolism, the metabolomes of whole bone, isolated cortical bone without marrow, and bone marrow have not been compared. We hypothesized that the metabolome of isolated cortical bone would be distinct from that of bone marrow and would reveal sex differences. Metabolite profiles from liquid chromatography–mass spectrometry (LC‐MS) of whole bone, isolated cortical bone, and bone marrow were generated from humeri from 20‐week‐old female C57Bl/6J mice. The cortical bone metabolomes were then compared for 20‐week‐old female and male C57Bl/6J mice. Femurs from male and female mice were evaluated for flexural material properties and were then categorized into bone strength groups. The metabolome of isolated cortical bone was distinct from both whole bone and bone marrow. We also found sex differences in the isolated cortical bone metabolome. Based on metabolite pathway analysis, females had higher lipid metabolism, and males had higher amino acid metabolism. High‐strength bones, regardless of sex, had greater tryptophan and purine metabolism. For males, high‐strength bones had upregulated nucleotide metabolism, whereas lower‐strength bones had greater pentose phosphate pathway metabolism. Because the higher‐strength groups (females compared with males, high‐strength males compared with lower‐strength males) had higher serum type I collagen cross‐linked C‐telopeptide (CTX1)/procollagen type 1 N propeptide (P1NP), we estimate that the metabolomic signature of bone strength in our study at least partially reflects differences in bone turnover. These data provide novel insight into bone bioenergetics and the sexual dimorphic nature of bone material properties in C57Bl/6 mice. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hope D. Welhaven
- Department of Chemistry & Biochemistry Montana State University Bozeman MT
- Molecular Biosciences Program Montana State University Bozeman MT
| | - Ghazal Vahidi
- Department of Mechanical & Industrial Engineering Montana State University Bozeman MT
| | - Seth T. Walk
- Department of Microbiology and Cell Biology Montana State University Bozeman MT
| | - Brian Bothner
- Department of Chemistry & Biochemistry Montana State University Bozeman MT
| | - Stephen A. Martin
- Translational Biomarkers Core Laboratory Montana State University Bozeman MT
| | - Chelsea M. Heveran
- Department of Mechanical & Industrial Engineering Montana State University Bozeman MT
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering Montana State University Bozeman MT
- Department of Microbiology and Cell Biology Montana State University Bozeman MT
| |
Collapse
|
29
|
Bartikoski BJ, de Oliveira MS, do Espírito Santo RC, dos Santos LP, dos Santos NG, Xavier RM. A Review of Metabolomic Profiling in Rheumatoid Arthritis: Bringing New Insights in Disease Pathogenesis, Treatment and Comorbidities. Metabolites 2022; 12:394. [PMID: 35629898 PMCID: PMC9146149 DOI: 10.3390/metabo12050394] [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: 03/08/2022] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Metabolomic analysis provides a wealth of information that can be predictive of distinctive phenotypes of pathogenic processes and has been applied to better understand disease development. Rheumatoid arthritis (RA) is an autoimmune disease with the establishment of chronic synovial inflammation that affects joints and peripheral tissues such as skeletal muscle and bone. There is a lack of useful disease biomarkers to track disease activity, drug response and follow-up in RA. In this review, we describe potential metabolic biomarkers that might be helpful in the study of RA pathogenesis, drug response and risk of comorbidities. TMAO (choline and trimethylamine oxide) and TCA (tricarboxylic acid) cycle products have been suggested to modulate metabolic profiles during the early stages of RA and are present systemically, which is a relevant characteristic for biomarkers. Moreover, the analysis of lipids such as cholesterol, FFAs and PUFAs may provide important information before disease onset to predict disease activity and treatment response. Regarding therapeutics, TNF inhibitors may increase the levels of tryptophan, valine, lysine, creatinine and alanine, whereas JAK/STAT inhibitors may modulate exclusively fatty acids. These observations indicate that different disease modifying antirheumatic drugs have specific metabolic profiles and can reveal differences between responders and non-responders. In terms of comorbidities, physical impairment represented by higher fatigue scores and muscle wasting has been associated with an increase in urea cycle, FFAs, tocopherols and BCAAs. In conclusion, synovial fluid, blood and urine samples from RA patients seem to provide critical information about the metabolic profile related to drug response, disease activity and comorbidities.
Collapse
Affiliation(s)
- Bárbara Jonson Bartikoski
- Laboratório de Doenças Autoimunes, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, RS, Brazil; (B.J.B.); (M.S.d.O.); (R.C.d.E.S.); (L.P.d.S.); (N.G.d.S.)
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
- Postgraduate Program in Medical Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2400, Porto Alegre 90035-003, RS, Brazil
| | - Marianne Schrader de Oliveira
- Laboratório de Doenças Autoimunes, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, RS, Brazil; (B.J.B.); (M.S.d.O.); (R.C.d.E.S.); (L.P.d.S.); (N.G.d.S.)
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
- Postgraduate Program in Medical Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2400, Porto Alegre 90035-003, RS, Brazil
| | - Rafaela Cavalheiro do Espírito Santo
- Laboratório de Doenças Autoimunes, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, RS, Brazil; (B.J.B.); (M.S.d.O.); (R.C.d.E.S.); (L.P.d.S.); (N.G.d.S.)
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
- Postgraduate Program in Medical Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2400, Porto Alegre 90035-003, RS, Brazil
| | - Leonardo Peterson dos Santos
- Laboratório de Doenças Autoimunes, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, RS, Brazil; (B.J.B.); (M.S.d.O.); (R.C.d.E.S.); (L.P.d.S.); (N.G.d.S.)
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
- Postgraduate Program in Medical Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2400, Porto Alegre 90035-003, RS, Brazil
| | - Natália Garcia dos Santos
- Laboratório de Doenças Autoimunes, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, RS, Brazil; (B.J.B.); (M.S.d.O.); (R.C.d.E.S.); (L.P.d.S.); (N.G.d.S.)
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
- Postgraduate Program in Biological Sciences: Pharmacology and Therapeutics, Barcelos 2400, Porto Alegre 90035-003, RS, Brazil
| | - Ricardo Machado Xavier
- Laboratório de Doenças Autoimunes, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-903, RS, Brazil; (B.J.B.); (M.S.d.O.); (R.C.d.E.S.); (L.P.d.S.); (N.G.d.S.)
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre 90035-903, RS, Brazil
- Postgraduate Program in Medical Science, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2400, Porto Alegre 90035-003, RS, Brazil
| |
Collapse
|
30
|
Fearon U, Hanlon MM, Floudas A, Veale DJ. Cellular metabolic adaptations in rheumatoid arthritis and their therapeutic implications. Nat Rev Rheumatol 2022; 18:398-414. [PMID: 35440762 DOI: 10.1038/s41584-022-00771-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Activation of endothelium and immune cells is fundamental to the initiation of autoimmune diseases such as rheumatoid arthritis (RA), and it results in trans-endothelial cell migration and synovial fibroblast proliferation, leading to joint destruction. In RA, the synovial microvasculature is highly dysregulated, resulting in inefficient oxygen perfusion to the synovium, which, along with the high metabolic demands of activated immune and stromal cells, leads to a profoundly hypoxic microenvironment. In inflamed joints, infiltrating immune cells and synovial resident cells have great requirements for energy and nutrients, and they adapt their metabolic profiles to generate sufficient energy to support their highly activated inflammatory states. This shift in metabolic capacity of synovial cells enables them to produce the essential building blocks to support their proliferation, activation and invasiveness. Furthermore, it results in the accumulation of metabolic intermediates and alteration of redox-sensitive pathways, affecting signalling pathways that further potentiate the inflammatory response. Importantly, the inflamed synovium is a multicellular tissue, with cells differing in their metabolic requirements depending on complex cell-cell interactions, nutrient supply, metabolic intermediates and transcriptional regulation. Therefore, understanding the complex interplay between metabolic and inflammatory pathways in synovial cells in RA will provide insight into the underlying mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland. .,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland.
| | - Megan M Hanlon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, TCD, Dublin, Ireland.,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
31
|
Berthelot JM, Bandiaky ON, Le Goff B, Amador G, Chaux AG, Soueidan A, Denis F. Another Look at the Contribution of Oral Microbiota to the Pathogenesis of Rheumatoid Arthritis: A Narrative Review. Microorganisms 2021; 10:59. [PMID: 35056507 PMCID: PMC8778040 DOI: 10.3390/microorganisms10010059] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022] Open
Abstract
Although autoimmunity contributes to rheumatoid arthritis (RA), several lines of evidence challenge the dogma that it is mainly an autoimmune disorder. As RA-associated human leukocyte antigens shape microbiomes and increase the risk of dysbiosis in mucosae, RA might rather be induced by epigenetic changes in long-lived synovial presenting cells, stressed by excessive translocations into joints of bacteria from the poorly cultivable gut, lung, or oral microbiota (in the same way as more pathogenic bacteria can lead to "reactive arthritis"). This narrative review (i) lists evidence supporting this scenario, including the identification of DNA from oral and gut microbiota in the RA synovium (but in also healthy synovia), and the possibility of translocation through blood, from mucosae to joints, of microbiota, either directly from the oral cavity or from the gut, following an increase of gut permeability worsened by migration within the gut of oral bacteria such as Porphyromonas gingivalis; (ii) suggests other methodologies for future works other than cross-sectional studies of periodontal microbiota in cohorts of patients with RA versus controls, namely, longitudinal studies of oral, gut, blood, and synovial microbiota combined with transcriptomic analyses of immune cells in individual patients at risk of RA, and in overt RA, before, during, and following flares of RA.
Collapse
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Unit, Nantes University Hospital, Place Alexis Ricordeau, CEDEX 01, 44093 Nantes, France; (J.-M.B.); (B.L.G.)
| | - Octave Nadile Bandiaky
- Division of Fixed Prosthodontics, University of Nantes, 1 Place Alexis Ricordeau, 44042 Nantes, France;
| | - Benoit Le Goff
- Rheumatology Unit, Nantes University Hospital, Place Alexis Ricordeau, CEDEX 01, 44093 Nantes, France; (J.-M.B.); (B.L.G.)
| | - Gilles Amador
- Department of Dental Public Health, Faculty of Dental Surgery, University of Nantes, 44093 Nantes, France;
- Nantes Teaching Hospital, 44000 Nantes, France;
| | - Anne-Gaelle Chaux
- Nantes Teaching Hospital, 44000 Nantes, France;
- Department of Oral Surgery, Faculty of Dental Surgery, University of Nantes, 44000 Nantes, France
| | - Assem Soueidan
- Department of Periodontology, Faculty of Dental Surgery, UIC 11, Rmes U1229, CHU de Nantes, 44000 Nantes, France;
| | - Frederic Denis
- Department of Dental Public Health, Faculty of Dental Surgery, University of Nantes, 44093 Nantes, France;
- Tours Teaching Hospital, 37000 Tours, France
| |
Collapse
|
32
|
Berthelot JM, Lioté F, Sibilia J. Tissue microbiota: a 'secondary-self', first target of autoimmunity? Joint Bone Spine 2021; 89:105337. [PMID: 34968748 DOI: 10.1016/j.jbspin.2021.105337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Department, Nantes University Hospital, Hôtel-Dieu, Place Alexis-Ricordeau, 44093, Nantes Cedex 01, France.
| | - Frédéric Lioté
- Rheumatology Department & Inserm UMR 1132 (centre Viggo Petersen), Hôpital Lariboisière, 2 rue Ambroise Paré, F-75010 Paris, France; Université de Paris, UFR de Médecine, F-75010 Paris, France
| | - Jean Sibilia
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France; RESO: Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, France; INSERM UMR_S1109, Université de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
33
|
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
|
34
|
Fajar JK, Ilmawan M, Mamada SS, Mutiawati E, Husnah M, Yusuf H, Nainu F, Sirinam S, Keam S, Ophinni Y, Rosiello F, Fahriani M, Rosa SGV. Global prevalence of persistent neuromuscular symptoms and the possible pathomechanisms in COVID-19 recovered individuals: A systematic review and meta-analysis. NARRA J 2021; 1:e48. [PMID: 38450213 PMCID: PMC10914045 DOI: 10.52225/narra.v1i3.48] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/26/2021] [Indexed: 02/05/2023]
Abstract
This study was conducted to determine the prevalence of prolonged neuromuscular symptoms, including fatigue, anosmia, headache, myalgia, and joint pain in COVID-19 survivors hospitalized with mild, moderate, or severe infections worldwide. The search was conducted up to January 30th, 2021 using three databases (PubMed, Scopus, and Web of Science) to identify potentially eligible studies. Data on study characteristics, follow-up characteristics, and severity of COVID-19 during hospitalization were collected in accordance with PRISMA guidelines. The Newcastle-Ottawa scale was used to assess the quality of relevant articles. The estimated prevalence of specific prolonged neuromuscular symptoms and the association between COVID-19 severity and occurrence of prolonged neuromuscular symptoms was analyzed wherever appropriate. Database search yielded 4,050 articles and 22 articles were included for meta-analysis. The estimated prevalence of prolonged fatigue was recorded in 21.2% (95%CI: 11.9%- 34.8%) of 3,730 COVID-19 survivors. Persistent anosmia was recorded in 239 of 2,600 COVID-19 survivors (9.7%, 95%CI: 6.1%-15.2%). In 84 out of 2,412 COVID-19 survivors (8.9%, 95%CI: 3.2%-22.6%), prolonged headache was observed. A total of 53 out of 1,125 COVID-19 patients (5.6%, 95%CI: 2.1%-14.2%) complained of persistent myalgia even after being discharged from the hospital. The prevalence of prolonged joint pain was in 15.4% (95%CI: 8.2%-27.2%) of subjects. Due to data scarcity on COVID-19 severity and prolonged neuromuscular symptoms, association analysis could not be conducted. Widespread concern regarding long-term impacts of COVID-19 was raised after several studies reported prolonged symptoms in COVID-19 survivors. Numerous theories have been proposed to address this concern; however, as the research on this pandemic is still ongoing, no explanation is definitive yet. Therefore, follow-up studies in COVID-19 survivors after recovery from COVID-19 are warranted to determine the pathogenesis of prolonged symptoms. PROSPERO registration: CRD42021242332.
Collapse
Affiliation(s)
- Jonny K Fajar
- Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | | | - Sukamto S Mamada
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar, Indonesia
| | - Endang Mutiawati
- Department of Neurology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Indonesia
| | - Milda Husnah
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Hanifah Yusuf
- Department of Pharmacology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar, Indonesia
| | - Salin Sirinam
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Synat Keam
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Youdiil Ophinni
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Harvard University, Cambridge, MA, USA
- Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Francesco Rosiello
- Department of Public Health and Infectious Disease, Sapienza-University of Rome, Rome, Italy
| | - Marhami Fahriani
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Sandro G V Rosa
- Diretoria de Patentes, Divisão De Farmácia - Instituto Nacional Da Propriedade Industrial, Rio de Janeiro, Brasil
- Programa De Pós-Graduação Em Ciências Aplicadas a Produtos Para Saúde, Faculdade De Farmácia, Universidade Federal Fluminense, Brasil
| |
Collapse
|
35
|
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
|
36
|
Atallah R, Gindlhuber J, Platzer W, Bärnthaler T, Tatzl E, Toller W, Strutz J, Rittchen S, Luschnig P, Birner-Gruenberger R, Wadsack C, Heinemann A. SUCNR1 Is Expressed in Human Placenta and Mediates Angiogenesis: Significance in Gestational Diabetes. Int J Mol Sci 2021; 22:12048. [PMID: 34769478 PMCID: PMC8585094 DOI: 10.3390/ijms222112048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Placental hypervascularization has been reported in pregnancy-related pathologies such as gestational diabetes mellitus (GDM). Nevertheless, the underlying causes behind this abnormality are not well understood. In this study, we addressed the expression of SUCNR1 (cognate succinate receptor) in human placental endothelial cells and hypothesized that the succinate-SUCNR1 axis might play a role in the placental hypervascularization reported in GDM. We measured significantly higher succinate levels in placental tissue lysates from women with GDM relative to matched controls. In parallel, SUCNR1 protein expression was upregulated in GDM tissue lysates as well as in isolated diabetic fetoplacental arterial endothelial cells (FpECAds). A positive correlation of SUCNR1 and vascular endothelial growth factor (VEGF) protein levels in tissue lysates indicated a potential link between the succinate-SUCNR1 axis and placental angiogenesis. In our in vitro experiments, succinate prompted hallmarks of angiogenesis in human umbilical vein endothelial cells (HUVECs) such as proliferation, migration and spheroid sprouting. These results were further validated in fetoplacental arterial endothelial cells (FpECAs), where succinate induced endothelial tube formation. VEGF gene expression was increased in response to succinate in both HUVECs and FpECAs. Yet, knockdown of SUCNR1 in HUVECs led to suppression of VEGF gene expression and abrogated the migratory ability and wound healing in response to succinate. In conclusion, our data underline SUCNR1 as a promising metabolic target in human placenta and as a potential driver of enhanced placental angiogenesis in GDM.
Collapse
MESH Headings
- Adult
- Case-Control Studies
- Cells, Cultured
- Diabetes, Gestational/genetics
- Diabetes, Gestational/metabolism
- Diabetes, Gestational/physiopathology
- Endothelium, Vascular/metabolism
- Female
- Human Umbilical Vein Endothelial Cells
- Humans
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Physiologic/genetics
- Placenta/blood supply
- Placenta/metabolism
- Pregnancy
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/physiology
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
- National Research Centre, Cairo 12622, Egypt
| | - Juergen Gindlhuber
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (J.G.); (R.B.-G.)
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria
| | - Wolfgang Platzer
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Thomas Bärnthaler
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Eva Tatzl
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria; (E.T.); (W.T.)
| | - Wolfgang Toller
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria; (E.T.); (W.T.)
| | - Jasmin Strutz
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (J.S.); (C.W.)
- Institute of Biomedical Science, Carinthia University of Applied Sciences, 9020 Klagenfurt, Austria
| | - Sonja Rittchen
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Petra Luschnig
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (J.G.); (R.B.-G.)
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (J.S.); (C.W.)
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| |
Collapse
|
37
|
Dubey D, Kumar S, Rawat A, Guleria A, Kumari R, Ahmed S, Singh R, Misra R, Kumar D. NMR-Based Metabolomics Revealed the Underlying Inflammatory Pathology in Reactive Arthritis Synovial Joints. J Proteome Res 2021; 20:5088-5102. [PMID: 34661415 DOI: 10.1021/acs.jproteome.1c00620] [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] [Indexed: 12/27/2022]
Abstract
Reactive arthritis (ReA) is an aseptic synovitis condition that often develops 2-4 weeks after a distant (extra-articular) infection with Chlamydia, Salmonella, Shigella, Campylobacter, and Yersinia species. The metabolic changes in the synovial fluid (SF) may serve as indicative markers to both improve the diagnostic accuracy and understand the underlying inflammatory pathology of ReA. With this aim, the metabolic profiles of SF collected from ReA (n = 58) and non-ReA, i.e., rheumatoid arthritis (RA, n = 21) and osteoarthritis (OA, n = 20) patients, respectively, were measured using NMR spectroscopy and compared using orthogonal partial least-squares discriminant analysis (OPLS-DA). The discriminatory metabolic features were further evaluated for their diagnostic potential using the receiver operating characteristic (ROC) curve analysis. Compared to RA, two (alanine and carnitine), and compared to OA, six (NAG, glutamate, glycerol, isoleucine, alanine, and glucose) metabolic features were identified as diagnostic biomarkers. We further demonstrated the impact of ReA synovitis condition on the serum metabolic profiles through performing a correlation analysis. The Pearson rank coefficient (r) was estimated for 38 metabolites (profiled in both SF and serum samples obtained in pair from ReA patients) and was found significantly positive for 71% of the metabolites (r ranging from 0.17 to 0.87).
Collapse
Affiliation(s)
- Durgesh Dubey
- Centre of Biomedical Research, Lucknow 226014, India.,Department of Clinical Immunology & Rheumatology, SGPGIMS, Lucknow 226014, India
| | - Sandeep Kumar
- Department of Clinical Immunology & Rheumatology, SGPGIMS, Lucknow 226014, India
| | - Atul Rawat
- Centre of Biomedical Research, Lucknow 226014, India
| | | | - Reena Kumari
- Department of Biochemistry, KGMU, Lucknow 226003, India
| | - Sakir Ahmed
- Department of Clinical Immunology & Rheumatology, SGPGIMS, Lucknow 226014, India.,Department of Clinical Immunology and Rheumatology, KIMS, Bhubaneswar 751024, India
| | - Rajeev Singh
- Regional Medical Research Center, Gorakhpur 273013, India
| | - Ramnath Misra
- Department of Clinical Immunology & Rheumatology, SGPGIMS, Lucknow 226014, India.,Department of Clinical Immunology and Rheumatology, KIMS, Bhubaneswar 751024, India
| | - Dinesh Kumar
- Centre of Biomedical Research, Lucknow 226014, India
| |
Collapse
|
38
|
Bhagavatham SKS, Khanchandani P, Kannan V, Potikuri D, Sridharan D, Pulukool SK, Naik AA, Dandamudi RB, Divi SM, Pargaonkar A, Ray R, Santha SSR, Seshagiri PB, Narasimhan K, Gumdal N, Sivaramakrishnan V. Adenosine deaminase modulates metabolic remodeling and orchestrates joint destruction in rheumatoid arthritis. Sci Rep 2021; 11:15129. [PMID: 34301999 PMCID: PMC8302689 DOI: 10.1038/s41598-021-94607-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic autoimmune disease associated with inflammation and joint remodeling. Adenosine deaminase (ADA), a risk factor in RA, degrades adenosine, an anti-inflammatory molecule, resulting in an inflammatory bias. We present an integrative analysis of clinical data, cytokines, serum metabolomics in RA patients and mechanistic studies on ADA-mediated effects on in vitro cell culture models. ADA activity differentiated patients into low and high ADA sets. The levels of the cytokines TNFα, IFNγ, IL-10, TGFβ and sRANKL were elevated in RA and more pronounced in high ADA sets. Serum metabolomic analysis shows altered metabolic pathways in RA which were distinct between low and high ADA sets. Comparative analysis with previous studies shows similar pathways are modulated by DMARDs and biologics. Random forest analysis distinguished RA from control by methyl-histidine and hydroxyisocaproic acid, while hexose-phosphate and fructose-6-phosphate distinguished high ADA from low ADA. The deregulated metabolic pathways of High ADA datasets significantly overlapped with high ADA expressing PBMCs GEO transcriptomics dataset. ADA induced the death of chondrocytes, synoviocyte proliferation, both inflammation in macrophages and their differentiation into osteoclasts and impaired differentiation of mesenchymal stem cells to osteoblasts and mineralization. PBMCs expressing elevated ADA had increased expression of cytokines and P2 receptors compared to synovial macrophages which has low expression of ADA. Our data demonstrates increased cytokine levels and distinct metabolic signatures of RA based on the ADA activity, suggests an important role for ADA in the pathophysiology of RA joints and as a potential marker and therapeutic target in RA patients.
Collapse
Affiliation(s)
- Sai Krishna Srimadh Bhagavatham
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Prakash Khanchandani
- grid.496668.30000 0004 1767 3076Department of Orthopedics, Sri Sathya Sai Institute of Higher Medical Sciences, PG, Puttaparthi, 515134 India
| | - Vishnu Kannan
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India ,grid.411552.60000 0004 1766 4022Present Address: Department of Botany/Biotechnology, CMS College, Kottayam, 686001 India
| | | | - Divya Sridharan
- grid.34980.360000 0001 0482 5067Molecular Reproduction and Developmental Genetics, Indian Institute of Science, Bengaluru, 560012 India
| | - Sujith Kumar Pulukool
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Ashwin Ashok Naik
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Rajesh Babu Dandamudi
- grid.444651.60000 0004 0496 6988Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, India ,Present Address: Phenomenex India, Hyderabad, Telangana 500084 India
| | - Sai Mangala Divi
- grid.496668.30000 0004 1767 3076Department of Biochemistry, Sri Sathya Sai Institute of Higher Medical Sciences, PG, Puttaparthi, 515134 India
| | - Ashish Pargaonkar
- grid.464737.50000 0004 1775 153XAgilent Technologies India Pvt Ltd, Bengaluru, 560048 India
| | - Rahul Ray
- grid.496668.30000 0004 1767 3076Department of Orthopedics, Sri Sathya Sai Institute of Higher Medical Sciences, PG, Puttaparthi, 515134 India
| | - Saibharath Simha Reddy Santha
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| | - Polani B. Seshagiri
- grid.34980.360000 0001 0482 5067Molecular Reproduction and Developmental Genetics, Indian Institute of Science, Bengaluru, 560012 India
| | - K. Narasimhan
- Sri Sathya Sai General Hospital, Puttaparthi, 515134 India
| | | | - Venketesh Sivaramakrishnan
- grid.444651.60000 0004 0496 6988Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Puttaparthi, 515134 India
| |
Collapse
|
39
|
Clayton SA, MacDonald L, Kurowska-Stolarska M, Clark AR. Mitochondria as Key Players in the Pathogenesis and Treatment of Rheumatoid Arthritis. Front Immunol 2021; 12:673916. [PMID: 33995417 PMCID: PMC8118696 DOI: 10.3389/fimmu.2021.673916] [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: 02/28/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022] Open
Abstract
Mitochondria are major energy-producing organelles that have central roles in cellular metabolism. They also act as important signalling hubs, and their dynamic regulation in response to stress signals helps to dictate the stress response of the cell. Rheumatoid arthritis is an inflammatory and autoimmune disease with high prevalence and complex aetiology. Mitochondrial activity affects differentiation, activation and survival of immune and non-immune cells that contribute to the pathogenesis of this disease. This review outlines what is known about the role of mitochondria in rheumatoid arthritis pathogenesis, and how current and future therapeutic strategies can function through modulation of mitochondrial activity. We also highlight areas of this topic that warrant further study. As producers of energy and of metabolites such as succinate and citrate, mitochondria help to shape the inflammatory phenotype of leukocytes during disease. Mitochondrial components can directly stimulate immune receptors by acting as damage-associated molecular patterns, which could represent an initiating factor for the development of sterile inflammation. Mitochondria are also an important source of intracellular reactive oxygen species, and facilitate the activation of the NLRP3 inflammasome, which produces cytokines linked to disease symptoms in rheumatoid arthritis. The fact that mitochondria contain their own genetic material renders them susceptible to mutation, which can propagate their dysfunction and immunostimulatory potential. Several drugs currently used for the treatment of rheumatoid arthritis regulate mitochondrial function either directly or indirectly. These actions contribute to their immunomodulatory functions, but can also lead to adverse effects. Metabolic and mitochondrial pathways are attractive targets for future anti-rheumatic drugs, however many questions still remain about the precise role of mitochondrial activity in different cell types in rheumatoid arthritis.
Collapse
Affiliation(s)
- Sally A Clayton
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Birmingham, United Kingdom
| | - Lucy MacDonald
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, United Kingdom.,Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Mariola Kurowska-Stolarska
- Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, United Kingdom.,Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Andrew R Clark
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Birmingham, United Kingdom
| |
Collapse
|
40
|
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
|
41
|
Kvacskay P, Yao N, Schnotz JH, Scarpone R, Carvalho RDA, Klika KD, Merkt W, Tretter T, Lorenz HM, Tykocinski LO. Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy? Arthritis Res Ther 2021; 23:56. [PMID: 33588937 PMCID: PMC7883459 DOI: 10.1186/s13075-021-02437-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/02/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A dysregulated glucose metabolism in synovial fibroblasts (SF) has been associated with their aggressive phenotype in rheumatoid arthritis (RA). Even though T helper (Th) cells are key effector cells in the propagation and exacerbation of synovitis in RA, little is known about their influence on the metabolism of SF. Thus, this study investigates the effect of Th cells on the glucose metabolism and phenotype of SF and how this is influenced by the blockade of cytokines, janus kinases (JAKs) and glycolysis. METHODS SF from patients with RA or osteoarthritis (OA) were cultured in the presence of a stable glucose isotopomer ([U-13C]-glucose) and stimulated with the conditioned media of activated Th cells (ThCM). Glucose consumption and lactate production were measured by proton nuclear magnetic resonance (1H NMR) spectroscopy. Cytokine secretion was quantified by ELISA. The expression of glycolytic enzymes was analysed by PCR, western blot and immunofluorescence. JAKs were blocked using either baricitinib or tofacitinib and glycolysis by using either 3-bromopyruvate or FX11. RESULTS Quiescent RASF produced significantly higher levels of lactate, interleukin (IL)-6 and matrix metalloproteinase (MMP) 3 than OASF. Stimulation by ThCM clearly changed the metabolic profile of both RASF and OASF by inducing a shift towards aerobic glycolysis with strongly increased lactate production together with a rise in IL-6 and MMP3 secretion. Interestingly, chronic stimulation of OASF by ThCM triggered an inflammatory phenotype with significantly increased glycolytic activity compared to unstimulated, singly stimulated or re-stimulated OASF. Finally, in contrast to cytokine-neutralizing biologics, inhibition of JAKs or glycolytic enzymes both significantly reduced lactate production and cytokine secretion by Th cell-stimulated SF. CONCLUSIONS Soluble mediators released by Th cells drive SF towards a glycolytic and pro-inflammatory phenotype. Targeting of JAKs or glycolytic enzymes both potently modulate SF's glucose metabolism and decrease the release of IL-6 and MMP3. Thus, manipulation of glycolytic pathways could represent a new therapeutic strategy to decrease the pro-inflammatory phenotype of SF.
Collapse
Affiliation(s)
- Peter Kvacskay
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Nina Yao
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Jürgen-Heinz Schnotz
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Roberta Scarpone
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Rui de Albuquerque Carvalho
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Karel D Klika
- Molecular Structure Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Merkt
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Theresa Tretter
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany
| | - Lars-Oliver Tykocinski
- Department of Medicine V, Division of Rheumatology, University of Heidelberg, INF 410, 69120, Heidelberg, Germany.
| |
Collapse
|
42
|
Mousavi MJ, Karami J, Aslani S, Tahmasebi MN, Vaziri AS, Jamshidi A, Farhadi E, Mahmoudi M. Transformation of fibroblast-like synoviocytes in rheumatoid arthritis; from a friend to foe. AUTO- IMMUNITY HIGHLIGHTS 2021; 12:3. [PMID: 33546769 PMCID: PMC7863458 DOI: 10.1186/s13317-020-00145-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Swelling and the progressive destruction of articular cartilage are major characteristics of rheumatoid arthritis (RA), a systemic autoimmune disease that directly affects the synovial joints and often causes severe disability in the affected positions. Recent studies have shown that type B synoviocytes, which are also called fibroblast-like synoviocytes (FLSs), as the most commonly and chiefly resident cells, play a crucial role in early-onset and disease progression by producing various mediators. During the pathogenesis of RA, the FLSs' phenotype is altered, and represent invasive behavior similar to that observed in tumor conditions. Modified and stressful microenvironment by FLSs leads to the recruitment of other immune cells and, eventually, pannus formation. The origins of this cancerous phenotype stem fundamentally from the significant metabolic changes in glucose, lipids, and oxygen metabolism pathways. Moreover, the genetic abnormalities and epigenetic alterations have recently been implicated in cancer-like behaviors of RA FLSs. In this review, we will focus on the mechanisms underlying the transformation of FLSs to a cancer-like phenotype during RA. A comprehensive understanding of these mechanisms may lead to devising more effective and targeted treatment strategies.
Collapse
Affiliation(s)
- Mohammad Javad Mousavi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Jafar Karami
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Arash Sharafat Vaziri
- Joint Reconstruction Reseach Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
43
|
Vitorino R, Guedes S, Vitorino C, Ferreira R, Amado F, Van Eyk JE. Elucidating Citrullination by Mass Spectrometry and Its Role in Disease Pathogenesis. J Proteome Res 2021; 20:38-48. [PMID: 32966086 PMCID: PMC11009872 DOI: 10.1021/acs.jproteome.0c00474] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This review focuses on discussing key mechanisms in disease pathogenesis mediated by the protein post-translational modification citrullination. These processes are discussed in depth in the context of complex diseases such as rheumatoid arthritis, cancer, central nervous system disorders, and cardiovascular disease. Additionally, a critical evaluation of challenges in laboratory detection of citrullination sites is also outlined. In this context, the role of mass spectrometry is discussed with a focus on contemporary techniques that offer promising options to detect the exact site of protein citrullination. Novel methods described in the paper have the potential to detect and quantify the occurrence of post-translational modification sites for diagnosis and therapeutic purposes with a high degree of specificity and sensitivity. Furthermore, they offer a much faster performance than traditional techniques making them ideal for large-scale experimentation.
Collapse
Affiliation(s)
- Rui Vitorino
- QOPNA & LAQV-REQUIMTE, Departamento de Qúimica, Universidade de Aveiro, Aveiro, Portugal; iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Sofia Guedes
- QOPNA & LAQV-REQUIMTE, Departamento de Qúimica, Universidade de Aveiro, Aveiro, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Rita Ferreira
- QOPNA & LAQV-REQUIMTE, Departamento de Qúimica, Universidade de Aveiro, Aveiro, Portugal
| | - Francisco Amado
- QOPNA & LAQV-REQUIMTE, Departamento de Qúimica, Universidade de Aveiro, Aveiro, Portugal
| | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinia Medical Center, Los Angeles, California, United States
| |
Collapse
|
44
|
Zinc and Cadmium in the Aetiology and Pathogenesis of Osteoarthritis and Rheumatoid Arthritis. Nutrients 2020; 13:nu13010053. [PMID: 33375344 PMCID: PMC7824316 DOI: 10.3390/nu13010053] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are inflammatory articular conditions with different aetiology, but both result in joint damage. The nutritionally essential metal zinc (Zn2+) and the non-essential metal cadmium (Cd2+) have roles in these arthritic diseases as effectors of the immune system, inflammation, and metabolism. Despite both metal ions being redox-inert in biology, they affect the redox balance. It has been known for decades that zinc decreases in the blood of RA patients. It is largely unknown, however, whether this change is only a manifestation of an acute phase response in inflammation or relates to altered availability of zinc in tissues and consequently requires changes of zinc in the diet. As a cofactor in over 3000 human proteins and as a signaling ion, zinc affects many pathways relevant for arthritic disease. How it affects the diseases is not just a question of zinc status, but also an issue of mutations in the many proteins that maintain cellular zinc homoeostasis, such as zinc transporters of the ZIP (Zrt-/Irt-like protein) and ZnT families and metallothioneins, and the multiple pathways that change the expression of these proteins. Cadmium interferes with zinc's functions and there is increased uptake under zinc deficiency. Remarkably, cadmium exposure through inhalation is now recognized in the activation of macrophages to a pro-inflammatory state and suggested as a trigger of a specific form of nodular RA. Here, we discuss how these metal ions participate in the genetic, metabolic, and environmental factors that lead to joint destruction. We conclude that both metal ions should be monitored routinely in arthritic disease and that there is untapped potential for prognosis and treatment.
Collapse
|
45
|
Ashrafian H, Sounderajah V, Glen R, Ebbels T, Blaise BJ, Kalra D, Kultima K, Spjuth O, Tenori L, Salek RM, Kale N, Haug K, Schober D, Rocca-Serra P, O'Donovan C, Steinbeck C, Cano I, de Atauri P, Cascante M. Metabolomics: The Stethoscope for the Twenty-First Century. Med Princ Pract 2020; 30:301-310. [PMID: 33271569 PMCID: PMC8436726 DOI: 10.1159/000513545] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/29/2020] [Indexed: 11/19/2022] Open
Abstract
Metabolomics encompasses the systematic identification and quantification of all metabolic products in the human body. This field could provide clinicians with novel sets of diagnostic biomarkers for disease states in addition to quantifying treatment response to medications at an individualized level. This literature review aims to highlight the technology underpinning metabolic profiling, identify potential applications of metabolomics in clinical practice, and discuss the translational challenges that the field faces. We searched PubMed, MEDLINE, and EMBASE for primary and secondary research articles regarding clinical applications of metabolomics. Metabolic profiling can be performed using mass spectrometry and nuclear magnetic resonance-based techniques using a variety of biological samples. This is carried out in vivo or in vitro following careful sample collection, preparation, and analysis. The potential clinical applications constitute disruptive innovations in their respective specialities, particularly oncology and metabolic medicine. Outstanding issues currently preventing widespread clinical use are scalability of data interpretation, standardization of sample handling practice, and e-infrastructure. Routine utilization of metabolomics at a patient and population level will constitute an integral part of future healthcare provision.
Collapse
Affiliation(s)
- Hutan Ashrafian
- Institute of Global Health Innovation and Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Viknesh Sounderajah
- Institute of Global Health Innovation and Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Robert Glen
- Institute of Global Health Innovation and Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Timothy Ebbels
- Institute of Global Health Innovation and Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Benjamin J. Blaise
- Institute of Global Health Innovation and Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Dipak Kalra
- Department of Medical Informatics and Statistics, University of Ghent, Ghent, Belgium
| | - Kim Kultima
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Leonardo Tenori
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Reza M. Salek
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Namrata Kale
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kenneth Haug
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Daniel Schober
- Department of Stress and Developmental Biology, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Philippe Rocca-Serra
- Department of Engineering Science, Oxford e-Research Centre, University of Oxford, Oxford, United Kingdom
| | - Claire O'Donovan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Christoph Steinbeck
- Institute for Inorganic and Analytical Chemistry, Friedrich-Schiller-University, Jena, Germany
| | - Isaac Cano
- Hospital Clinic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Pedro de Atauri
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona and CIBERHD (CIBER de Enfermedades hepáticas y digestivas), Barcelona, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona and CIBERHD (CIBER de Enfermedades hepáticas y digestivas), Barcelona, Spain
| |
Collapse
|
46
|
Jiang B, Hu L, Zhang X, Zhang H, Zhang F, Chen L, Li Z, Zhao X, Xue C, Jiang X. Uncovering proteome variations of differently heat-treated sea cucumber (Apostichopus japonicus) by label-free mass spectrometry. Food Chem 2020; 344:128575. [PMID: 33191009 DOI: 10.1016/j.foodchem.2020.128575] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 10/08/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023]
Abstract
The effects of heat treatment on the proteome of Apostichopus japonicus have been evaluated using label-free quantitative proteomics by ultrahigh performance liquid chromatography-quadrupole/time of flight (UHPLC-Q/TOF) mass spectrometry with sequential window acquisition of all the theoretical fragment ion (SWATH) acquisition mode. Chemometric tools are integrated to reveal proteomic changes by mining the protein quantitation data from fresh and differently heat-treated samples. SWATH allows the quantitation of 548 proteins, of which 24 proteins are significantly sensitive to heat treatment and 13 proteins vary significantly responding to different heat procedures (boiling, steaming, and microwave heating), and 5 of them are sharing proteins. Gene ontology (GO) annotation of the differentiating proteins highlights most of them are relevant to molecular functions. The results can be favorable to evaluate the effects of heat treatment on the nutrition and function of processed sea cucumbers and facilitate the selection of an optimal thermal treatment.
Collapse
Affiliation(s)
- Bingxue Jiang
- College of Food Science and Technology, Ocean University of China, No. 5 Yu Shan Road, Qingdao, Shandong Province 266003, PR China
| | - Lingping Hu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, PR China
| | - Xiaomei Zhang
- Technology Center of Qingdao Customs District, No.70 Qutangxia Road, Qingdao, Shandong Province 266002, PR China
| | - Hongwei Zhang
- Technology Center of Qingdao Customs District, No.70 Qutangxia Road, Qingdao, Shandong Province 266002, PR China.
| | - Feng Zhang
- Chinese Academy of Inspection & Quarantine, Beijing 100176, PR China
| | - Lipin Chen
- College of Food Science and Technology, Ocean University of China, No. 5 Yu Shan Road, Qingdao, Shandong Province 266003, PR China
| | - Zhaojie Li
- College of Food Science and Technology, Ocean University of China, No. 5 Yu Shan Road, Qingdao, Shandong Province 266003, PR China
| | - Xue Zhao
- College of Food Science and Technology, Ocean University of China, No. 5 Yu Shan Road, Qingdao, Shandong Province 266003, PR China
| | - Changhu Xue
- College of Food Science and Technology, Ocean University of China, No. 5 Yu Shan Road, Qingdao, Shandong Province 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong Province 266237, PR China
| | - Xiaoming Jiang
- College of Food Science and Technology, Ocean University of China, No. 5 Yu Shan Road, Qingdao, Shandong Province 266003, PR China.
| |
Collapse
|
47
|
Velcicky J, Wilcken R, Cotesta S, Janser P, Schlapbach A, Wagner T, Piechon P, Villard F, Bouhelal R, Piller F, Harlfinger S, Stringer R, Fehlmann D, Kaupmann K, Littlewood-Evans A, Haffke M, Gommermann N. Discovery and Optimization of Novel SUCNR1 Inhibitors: Design of Zwitterionic Derivatives with a Salt Bridge for the Improvement of Oral Exposure. J Med Chem 2020; 63:9856-9875. [DOI: 10.1021/acs.jmedchem.0c01020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Juraj Velcicky
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Rainer Wilcken
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Simona Cotesta
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Philipp Janser
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Achim Schlapbach
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Trixie Wagner
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Philippe Piechon
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Frederic Villard
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Rochdi Bouhelal
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Fabian Piller
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Rowan Stringer
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Klemens Kaupmann
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | | | - Matthias Haffke
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Nina Gommermann
- Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
48
|
Akhbari P, Karamchandani U, Jaggard MKJ, Graça G, Bhattacharya R, Lindon JC, Williams HRT, Gupte CM. Can joint fluid metabolic profiling (or "metabonomics") reveal biomarkers for osteoarthritis and inflammatory joint disease?: A systematic review. Bone Joint Res 2020; 9:108-119. [PMID: 32435463 PMCID: PMC7229296 DOI: 10.1302/2046-3758.93.bjr-2019-0167.r1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aims Metabolic profiling is a top-down method of analysis looking at metabolites, which are the intermediate or end products of various cellular pathways. Our primary objective was to perform a systematic review of the published literature to identify metabolites in human synovial fluid (HSF), which have been categorized by metabolic profiling techniques. A secondary objective was to identify any metabolites that may represent potential biomarkers of orthopaedic disease processes. Methods A systematic review was conducted in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines using the MEDLINE, Embase, PubMed, and Cochrane databases. Studies included were case series, case control series, and cohort studies looking specifically at HSF. Results The primary analysis, which pooled the results from 17 published studies and four meeting abstracts, identified over 200 metabolites. Seven of these studies (six published studies, one meeting abstract) had asymptomatic control groups and collectively suggested 26 putative biomarkers in osteoarthritis, inflammatory arthropathies, and trauma. These can broadly be categorized into amino acids plus related metabolites, fatty acids, ketones, and sugars. Conclusion The role of metabolic profiling in orthopaedics is fast evolving with many metabolites already identified in a variety of pathologies. However, these results need to be interpreted with caution due to the presence of multiple confounding factors in many of the studies. Future research should include largescale epidemiological metabolic profiling studies incorporating various confounding factors with appropriate statistical analysis to account for multiple testing of the data. Cite this article:Bone Joint Res. 2020;9(3):108–119.
Collapse
Affiliation(s)
- Pouya Akhbari
- Department of Orthopaedics & Trauma, Imperial College Healthcare NHS Trust, London, UK
| | | | - Matthew K J Jaggard
- Department of Orthopaedics & Trauma, Imperial College Healthcare NHS Trust, London, UK
| | - Goncalo Graça
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Rajarshi Bhattacharya
- Department of Orthopaedics & Trauma, Imperial College Healthcare NHS Trust, London, UK
| | - John C Lindon
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Horace R T Williams
- Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Chinmay M Gupte
- Department of Surgery and Cancer, Imperial College London, and Department of Orthopaedics & Trauma, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
49
|
Liu P, Wang J, Wen W, Pan T, Chen H, Fu Y, Wang F, Huang JH, Xu S. Cinnamaldehyde suppresses NLRP3 derived IL-1β via activating succinate/HIF-1 in rheumatoid arthritis rats. Int Immunopharmacol 2020; 84:106570. [PMID: 32413739 DOI: 10.1016/j.intimp.2020.106570] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/18/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Cinnamaldehyde (CA) is an essential component of cinnamon (Cinnamomum cassia Presland), which is often used as a flavoring condiment in beverages, pastries, perfumes, etc. Cinnamon is also used as herbal medicine in China and Southeast Asia to treat rheumatoid arthritis. However, the molecular mechanism is unclear. In this study, we aim to investigate its anti-inflammatory effects against Rheumatoid arthritis (RA) using activated macrophages (Raw246.7) in vitro and adjuvant arthritis rats (AA) in vivo. The results demonstrated that CA significantly reduced synovial inflammation in AA rats, possibly due to suppression of the expressions of pro-inflammatory cytokines, especially the IL-1β. Further investigation found that CA also suppressed the activity of HIF-1α by inhibiting the accumulation of succinate in cytoplasm. As we know, the reduction of HIF-1α nucleation slows down IL-1β production, because HIF-1α activates the expression of NLRP3, which is involved in the assembly of inflammasome and processing of IL-1β. In addition, CA also inhibited the expression of the succinate receptor GPR91, which in turn inhibited the activation of HIF-1α. In conclusions, our results suggested that CA might be a potential therapeutic compound to relieve rheumatoid arthritis progress by suppressing IL-1β through modulating succinate/HIF-1α axis and inhibition of NLRP3.
Collapse
Affiliation(s)
- Panwang Liu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Jie Wang
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Wen Wen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ting Pan
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Huan Chen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ying Fu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Fushun Wang
- Institute of Brain and Psyhological Sciences, Sichuan Normal University, Chengdu, Sichuan 610000, China
| | - Jason H Huang
- Department of Neurosurgery, Balor Scott & White Health Science Center, Temple, TX, United States; Department of Surgery, Texas A&M University College of Medicine, Temple, TX 79409, United States
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| |
Collapse
|
50
|
Burja B, Mertelj T, Frank-Bertoncelj M. Hi- JAKi-ng Synovial Fibroblasts in Inflammatory Arthritis With JAK Inhibitors. Front Med (Lausanne) 2020; 7:124. [PMID: 32432116 PMCID: PMC7214667 DOI: 10.3389/fmed.2020.00124] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022] Open
Abstract
The Janus kinase (JAK)-Signal transducer and activator of transcription (STAT) pathway is one of the central signaling hubs in inflammatory, immune and cancer cells. Inhibiting the JAK-STAT pathway with JAK inhibitors (jakinibs) constitutes an important therapeutic strategy in cancer and chronic inflammatory diseases like rheumatoid arthritis (RA). FDA has approved different jakinibs for the treatment of RA, including tofacitinib, baricitinib and upadacitinib, and several jakinibs are being tested in clinical trials. Here, we reviewed published studies of jakinib effects on resolving synovial pathology in inflammatory arthritis. We discussed the results of jakinibs on structural joint damage in clinical trials and explored the effects of jakinibs across different in vitro, ex vivo, and in vivo synovial experimental models. We delved rigorously into experimental designs of in vitro fibroblast studies, deconvoluted jakinib efficacy in synovial fibroblasts across diverse experimental conditions and discussed their translatability in vivo. Synovial fibroblasts can readily activate the JAK-STAT signaling pathway in response to cytokine stimulation. We highlighted rather limited effects of jakinibs on the in vitro cultured synovial fibroblasts and inferred that direct and indirect (immune cell-dependent) actions of jakinibs are required to curb the fibroblast pathology in vivo. These actions have not been mimicked optimally in current in vitro experimental designs, where inflammatory stimuli do not naturally clear out with treatment as they do in vivo. While summarizing the broad knowledge of synovial jakinib effects, our review uniquely challenges future study designs to better mimick the jakinib actions in broader cell communities, as occurring in vivo in the inflamed synovium. This can deepen our understanding of collective synovial activities of jakinibs and their therapeutic limitations, thereby fostering jakinib development in arthritis.
Collapse
Affiliation(s)
- Blaž Burja
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland.,Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tonja Mertelj
- Center of Experimental Rheumatology, University Hospital Zurich, Schlieren, Switzerland
| | | |
Collapse
|