1
|
Delgado Dolset MI, Pablo-Torres C, Contreras N, Couto-Rodríguez A, Escolar-Peña A, Graña-Castro O, Izquierdo E, López-Rodríguez JC, Macías-Camero A, Pérez-Gordo M, Villaseñor A, Zubeldia-Varela E, Barber D, Escribese MM. Severe Allergy as a Chronic Inflammatory Condition From a Systems Biology Perspective. Clin Exp Allergy 2024; 54:550-584. [PMID: 38938054 DOI: 10.1111/cea.14517] [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: 01/17/2024] [Revised: 05/14/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
Persistent and unresolved inflammation is a common underlying factor observed in several and seemingly unrelated human diseases, including cardiovascular and neurodegenerative diseases. Particularly, in atopic conditions, acute inflammatory responses such as those triggered by insect venom, food or drug allergies possess also a life-threatening potential. However, respiratory allergies predominantly exhibit late immune responses associated with chronic inflammation, that can eventually progress into a severe phenotype displaying similar features as those observed in other chronic inflammatory diseases, as is the case of uncontrolled severe asthma. This review aims to explore the different facets and systems involved in chronic allergic inflammation, including processes such as tissue remodelling and immune cell dysregulation, as well as genetic, metabolic and microbiota alterations, which are common to other inflammatory conditions. Our goal here was to deepen on the understanding of an entangled disease as is chronic allergic inflammation and expose potential avenues for the development of better diagnostic and intervention strategies.
Collapse
Affiliation(s)
- M I Delgado Dolset
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - C Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - N Contreras
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Couto-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Escolar-Peña
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - O Graña-Castro
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Izquierdo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - J C López-Rodríguez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Macías-Camero
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - A Villaseñor
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - E Zubeldia-Varela
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - D Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| | - M M Escribese
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada-Nemesio Díez (IMMA-ND), Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Spain
| |
Collapse
|
2
|
Masuoka S, Nishio J, Yamada S, Saito K, Kaneko K, Kaburaki M, Tanaka N, Sato H, Muraoka S, Kawazoe M, Mizutani S, Furukawa K, Ishii-Watabe A, Kawai S, Saito Y, Nanki T. Relationship Between the Lipidome Profile and Disease Activity in Patients with Rheumatoid Arthritis. Inflammation 2024; 47:1444-1458. [PMID: 38401020 DOI: 10.1007/s10753-024-01986-8] [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: 09/08/2023] [Revised: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 02/26/2024]
Abstract
Lipid mediators have been suggested to play important roles in the pathogenesis of rheumatoid arthritis (RA). Lipidomics has recently allowed for the comprehensive analysis of lipids and has revealed the potential of lipids as biomarkers for the early diagnosis of RA and prediction of therapeutic responses. However, the relationship between disease activity and the lipid profile in RA remains unclear. In the present study, we performed a plasma lipidomic analysis of 278 patients with RA during treatment and examined relationships with disease activity using the Disease Activity Score in 28 joints (DAS28)-erythrocyte sedimentation rate (ESR). In all patients, five lipids positively correlated and seven lipids negatively correlated with DAS28-ESR. Stearic acid [FA(18:0)] (r = -0.45) and palmitic acid [FA(16:0)] (r = -0.38) showed strong negative correlations. After adjustments for age, body mass index (BMI), and medications, stearic acid, palmitic acid, bilirubin, and lysophosphatidylcholines negatively correlated with disease activity. Stearic acid inhibited osteoclast differentiation from peripheral blood monocytes in in vitro experiments, suggesting its contribution to RA disease activity by affecting bone metabolism. These results indicate that the lipid profile correlates with the disease activity of RA and also that some lipids may be involved in the pathogenesis of RA.
Collapse
Affiliation(s)
- Shotaro Masuoka
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Junko Nishio
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
- Department of Immunopathology and Immunoregulation, Toho University School of Medicine, Tokyo, Japan
| | - Soichi Yamada
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Kosuke Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Kaichi Kaneko
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Makoto Kaburaki
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Nahoko Tanaka
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Hiroshi Sato
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Sei Muraoka
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Mai Kawazoe
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Satoshi Mizutani
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Karin Furukawa
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Shinichi Kawai
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan
- Department of Inflammation and Pain Control Research, Toho University School of Medicine, Tokyo, Japan
| | - Yoshiro Saito
- Division of Medicinal Safety Science, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Toshihiro Nanki
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, 6-11-1 Omori-Nishi, Ota-Ku, Tokyo, 143-8541, Japan.
| |
Collapse
|
3
|
Sharma SD, Bluett J. Towards Personalized Medicine in Rheumatoid Arthritis. Open Access Rheumatol 2024; 16:89-114. [PMID: 38779469 PMCID: PMC11110814 DOI: 10.2147/oarrr.s372610] [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: 01/04/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, incurable, multisystem, inflammatory disease characterized by synovitis and extra-articular features. Although several advanced therapies targeting inflammatory mechanisms underlying the disease are available, no advanced therapy is universally effective. Therefore, a ceiling of treatment response is currently accepted where no advanced therapy is superior to another. The current challenge for medical research is the discovery and integration of predictive markers of drug response that can be used to personalize medicine so that the patient is started on "the right drug at the right time". This review article summarizes our current understanding of predicting response to anti-rheumatic drugs in RA, obstacles impeding the development of personalized medicine approaches and future research priorities to overcome these barriers.
Collapse
Affiliation(s)
- Seema D Sharma
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - James Bluett
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Yu Y, Zhang B, Jiang X, Cui Y, Luo H, Stergiadis S, Wang B. Exploring the metabolomic landscape: Perilla frutescens as a promising enhancer of production, flavor, and nutrition in Tan lamb meat. Meat Sci 2024; 209:109419. [PMID: 38154372 DOI: 10.1016/j.meatsci.2023.109419] [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: 07/18/2023] [Revised: 11/20/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
Addressing health-related concerns linked to the metabolite profile of lamb meat has become paramount, in line with the growing demand for enhanced flavor and taste. We examined the impact of Perilla frutescens seeds on Tan lamb growth, carcass traits, and metabolite profiles. Three diets were employed: a low-concentrate group (LC), a high-concentrate group (HC), and a PFS group (the LC diet supplemented with 3% Perilla frutescens seeds) on a dry matter basis. Forty-five male Tan-lambs (approximately six months) with similar body weights (25.1 kg ± 1.12 SD) were randomly assigned to one of these three groups for 84-day feeding, including an initial 14-day adjustment phase. The supplementation of PFS resulted in increased average daily gain (P < 0.01) and improved carcass quality and meat color (P < 0.05). Additionally, it led to an enhancement in omega-3 polyunsaturated fatty acids (P < 0.05) and a reduction in the omega-6/omega-3 ratio (P < 0.05). Using gas chromatography-mass spectrometry, 369 volatile compounds were identified with enhanced levels of acetaldehyde and 1,2,4-trimethyl-benzene associated with PFS (P < 0.05). Among the 807 compounds identified by ultra-high performance liquid chromatography-mass spectrometry, there were 66 significantly differential compounds (P < 0.05), including 43 hydrophilic metabolites and 23 lipids. PFS supplementation led to significant alterations in 66 metabolites, with three metabolites including 2,5-diisopropyl-3-methylphenol, 3-hydroxydecanoic acid, and lysophosphatidylcholine (15:0) emerging as potential PFS-related biomarkers. The study indicates that PFS supplementation can enhance Tan-lamb growth, feed efficiency, and meat quality, potentially providing lamb meat with improved flavor and nutritional characteristics.
Collapse
Affiliation(s)
- Yue Yu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Boyan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xianzhe Jiang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yimeng Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Sokratis Stergiadis
- University of Reading, School of Agriculture, Policy and Development, Department of Animal Sciences, Reading RG6 6EU, United Kingdom
| | - Bing Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
5
|
Zhang L, Jiang S, Guan Z, Huang J, Yin Z, Tan G, Wang Y, Zhao Z, Huang M, Jin J. Effect of Salvia miltiorrhiza Bunge extracts on improving the efficacy and reducing the toxicity of Tripterygium wilfordii polyglycosides in the treatment of rheumatoid arthritis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116782. [PMID: 37321427 DOI: 10.1016/j.jep.2023.116782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii polyglycosides (TWP), extracted from the traditional Chinese herb Tripterygium wilfordii, has been widely used in the treatment of rheumatoid arthritis (RA). However, the toxicity of TWP to a variety of organs such as liver, kidney and testis greatly limits its clinical application. Salvia miltiorrhiza Bunge is often used in the treatment of RA due to its blood circulation promoting, stasis resolving, and anti-inflammatory effects. Salvia miltiorrhiza Bunge has also been reported to possess multiple organ protective effects. AIM OF THE STUDY To investigate the influences of two main components of Salviorrhiza miltiorrhiza Bunge, hydrophilic salvianolic acids (SA) and lipophilic tanshinones (Tan), on the efficacy and toxicity of TWP in treating RA and to explore the underlying mechanisms. MATERIALS AND METHODS SA and Tan were extracted from Salvia miltiorrhiza Bunge and the extracts were quantitated by HPLC and identified by UPLC-Q/TOF-MS. Then, a collagen-induced arthritis (CIA) rat model was established using bovine type II collagen (CII) and incomplete Freund's adjuvant (IFA). CIA rats were treated with TWP and/or SA/Tan. After 21 days of continuous treatment, arthritis symptoms and organs toxicity were evaluated. Meanwhile, serum metabolomics were investigated by the UPLC-Q/TOF-MS to understand the underlying mechanism. RESULTS SA and Tan extracts could significantly alleviate arthritis symptoms in CIA rats and decrease the serum levels of inflammatory factors TNF-α, IL-1β and IL-6 when combined with TWP. Meanwhile, both extracts alleviated injury of liver, kidney and testis caused by TWP, and the hydrophilic extract SA was superior. Moreover, a total of 38 endogenous differential metabolites were identified between the CIA model group and the TWP group, among which 33 metabolites were significantly recovered after the combination of SA or Tan. Metabolic pathway analysis showed that SA and Tan can affect metabolic pathways including linoleic acid metabolism, glycerophospholipid metabolism, sphingolipid metabolism and steroid biosynthesis metabolism pathway. CONCLUSIONS Our findings indicated for the first time that two Salviorrhiza miltiorrhiza Bunge extracts could improve the efficacy and reduce the toxicity of TWP in the treatment of RA by adjusting metabolic pathways, and the hydrophilic extract SA was superior.
Collapse
Affiliation(s)
- Lei Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zehao Guan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Junyuan Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhaokun Yin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guoyao Tan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuanyuan Wang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
6
|
Chaplin H, Bosworth A, Simpson C, Wilkins K, Meehan J, Nikiphorou E, Moss-Morris R, Lempp H, Norton S. Refractory inflammatory arthritis definition and model generated through patient and multi-disciplinary professional modified Delphi process. PLoS One 2023; 18:e0289760. [PMID: 37556424 PMCID: PMC10411820 DOI: 10.1371/journal.pone.0289760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023] Open
Abstract
OBJECTIVE Various definitions have been proposed for Refractory Disease in people with Rheumatoid Arthritis; however, none were generated for Polyarticular Juvenile Idiopathic Arthritis or involving adult and paediatric multidisciplinary healthcare professionals and patients. The study aim is to redefine Refractory Disease, using Delphi methodology. METHODS Three rounds of surveys (one nominal group and two online (2019-2020)) to achieve consensus using a predetermined cut-off were conducted voting on: a) name, b) treatment and inflammation, c) symptoms and impact domains, and d) rating of individual components within domains. Theoretical application of the definition was conducted through a scoping exercise. RESULTS Votes were collected across three rounds from Patients, Researchers and nine multi-disciplinary healthcare professional groups (n = 106). Refractory Inflammatory Arthritis was the most popular name. Regarding treatment and inflammation, these were voted to be kept broad rather than specifying numbers/cut-offs. From 10 domains identified to capture symptoms and disease impact, six domains reached consensus for inclusion: 1) Disease Activity, 2) Joint Involvement, 3) Pain, 4) Fatigue, 5) Functioning and Quality of Life, and 6) Disease-Modifying Anti-Rheumatic Drug Experiences. Within these domains, 18 components, from an initial pool (n = 73), were identified as related and important to capture multi-faceted presentation of Refractory Inflammatory Arthritis, specifically in Rheumatoid Arthritis and Polyarticular Juvenile Idiopathic Arthritis. Feasibility of the revised definition was established (2022-2023) with good utility as was applied to 82% of datasets (n = 61) incorporating 20 outcome measures, with two further measures added to increase its utility and coverage of Pain and Fatigue. CONCLUSION Refractory Inflammatory Arthritis has been found to be broader than not achieving low disease activity, with wider biopsychosocial components and factors incorporating Persistent Inflammation or Symptoms identified as important. This definition needs further refinement to assess utility as a classification tool to identify patients with unmet needs.
Collapse
Affiliation(s)
- Hema Chaplin
- Health Psychology Section, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Ailsa Bosworth
- National Rheumatoid Arthritis Society, White Waltham, United Kingdom
| | - Carol Simpson
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | - Kate Wilkins
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | - Jessica Meehan
- Health Psychology Section, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | - Rona Moss-Morris
- Health Psychology Section, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Heidi Lempp
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | - Sam Norton
- Health Psychology Section, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| |
Collapse
|
7
|
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
|
8
|
Ravaei A, Pulsatelli L, Assirelli E, Ciaffi J, Meliconi R, Salvarani C, Govoni M, Rubini M. MTHFR c.665C>T and c.1298A>C Polymorphisms in Tailoring Personalized Anti-TNF-α Therapy for Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24044110. [PMID: 36835522 PMCID: PMC9962934 DOI: 10.3390/ijms24044110] [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: 02/02/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease with a prevalence of 1%. Currently, RA treatment aims to achieve low disease activity or remission. Failure to achieve this goal causes disease progression with a poor prognosis. When treatment with first-line drugs fails, treatment with tumor necrosis factor-α (TNF-α) inhibitors may be prescribed to which many patients do not respond adequately, making the identification of response markers urgent. This study investigated the association of two RA-related genetic polymorphisms, c.665C>T (historically referred to as C677T) and c.1298A>C, in the MTHFR gene as response markers to an anti-TNF-α therapy. A total of 81 patients were enrolled, 60% of whom responded to the therapy. Analyses showed that both polymorphisms were associated with a response to therapy in an allele dose-dependent manner. The association for c.665C>T was significant for a rare genotype (p = 0.01). However, the observed opposite trend of association for c.1298A>C was not significant. An analysis revealed that c.1298A>C, unlike c.665C>T, was also significantly associated with the drug type (p = 0.032). Our preliminary results showed that the genetic polymorphisms in the MTHFR gene were associated with a response to anti-TNF-α therapy, with a potential significance for the anti-TNF-α drug type. This evidence suggests a role for one-carbon metabolism in anti-TNF-α drug efficacy and contributes to further personalized RA interventions.
Collapse
Affiliation(s)
- Amin Ravaei
- Medical Genetics Laboratory, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Lia Pulsatelli
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Elisa Assirelli
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Jacopo Ciaffi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Riccardo Meliconi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Carlo Salvarani
- Division of Rheumatology, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
- University-Hospital of Modena, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Marcello Govoni
- Section of Hematology and Rheumatology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Rheumatology Unit, Sant’Anna University Hospital, 44124 Ferrara, Italy
| | - Michele Rubini
- Medical Genetics Laboratory, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
- University Center for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-974473
| |
Collapse
|
9
|
Zhu J, Wang T, Lin Y, Xiong M, Chen J, Jian C, Zhang J, Xie H, Zeng F, Huang Q, Su J, Zhao Y, Li S, Zeng F. The change of plasma metabolic profile and gut microbiome dysbiosis in patients with rheumatoid arthritis. Front Microbiol 2022; 13:931431. [PMID: 36329847 PMCID: PMC9623673 DOI: 10.3389/fmicb.2022.931431] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/19/2022] [Indexed: 11/14/2022] Open
Abstract
Objective Rheumatoid arthritis (RA) is a chronic inflammatory joint disease, which is associated with progressive disability, systemic complications, and early death. But its etiology and pathogenesis are not fully understood. We aimed to investigate the alterations in plasma metabolite profiles, gut bacteria, and fungi and their role of them in the pathogenesis of RA. Methods Metabolomics profiling of plasma from 363 participants including RA (n = 244), systemic lupus erythematosus (SLE, n = 50), and healthy control (HC, n = 69) were performed using the ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry. The differentially expressed metabolites were selected among groups and used to explore important metabolic pathways. Gut microbial diversity analysis was performed by 16S rRNA sequencing and ITS sequencing (RA = 195, HC = 269), and the specific microbial floras were identified afterward. The diagnosis models were established based on significant differential metabolites and microbial floras, respectively. Results There were 63 differential metabolites discovered between RA and HC groups, mainly significantly enriched in the arginine and proline metabolism, glycine, serine, and threonine metabolism, and glycerophospholipid metabolism between RA and HC groups. The core differential metabolites included L-arginine, creatine, D-proline, ornithine, choline, betaine, L-threonine, LysoPC (18:0), phosphorylcholine, and glycerophosphocholine. The L-arginine and phosphorylcholine were increased in the RA group. The AUC of the predictive model was 0.992, based on the combination of the 10 differential metabolites. Compared with the SLE group, 23 metabolites increased and 61 metabolites decreased in the RA group. However, no significant metabolic pathways were enriched between RA and SLE groups. On the genus level, a total of 117 differential bacteria genera and 531 differential fungal genera were identified between RA and HC groups. The results indicated that three bacteria genera (Eubacterium_hallii_group, Escherichia-Shigella, Streptococcus) and two fungal genera (Candida and Debaryomyces) significantly increased in RA patients. The AUC was 0.80 based on a combination of six differential bacterial genera and the AUC was 0.812 based on a combination of seven differential fungal genera. Functional predictive analysis displayed that differential bacterial and differential fungus both were associated with KEGG pathways involving superpathway of L-serine and glycine biosynthesis I, arginine, ornithine, and proline interconversion. Conclusion The plasma metabolism profile and gut microbe profile changed markedly in RA. The glycine, serine, and threonine metabolism and arginine and proline metabolism played an important role in RA.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tingting Wang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Yifei Lin
- Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, China
| | - Minghao Xiong
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | | | - Congcong Jian
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhang
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Huanhuan Xie
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Fanwei Zeng
- Sichuan Province Orthopaedic Hospital, Chengdu, China
| | - Qian Huang
- Dazhou Vocational and Technical College, Dazhou, China
| | - Jiang Su
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Clinical Institute of Inflammation and Immunology, Sichuan University, Chengdu, China
- *Correspondence: Yi Zhao,
| | - Shilin Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
- Shilin Li,
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
- Fanxin Zeng,
| |
Collapse
|
10
|
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] [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
- *Correspondence: Shicheng Guo, ; Dongyi He,
| | - 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
- *Correspondence: Shicheng Guo, ; Dongyi He,
| |
Collapse
|
11
|
Gene Ontology Analysis Highlights Biological Processes Influencing Non-Response to Anti-TNF Therapy in Rheumatoid Arthritis. Biomedicines 2022; 10:biomedicines10081808. [PMID: 36009355 PMCID: PMC9404936 DOI: 10.3390/biomedicines10081808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
Anti-TNF therapy has significantly improved disease control in rheumatoid arthritis, but a fraction of rheumatoid arthritis patients do not respond to anti-TNF therapy or lose response over time. Moreover, the mechanisms underlying non-response to anti-TNF therapy remain largely unknown. To date, many single biomarkers of response to anti-TNF therapy have been published but they have not yet been analyzed as a system of interacting nodes. The aim of our study is to systematically elucidate the biological processes underlying non-response to anti-TNF therapy in rheumatoid arthritis using the gene ontologies of previously published predictive biomarkers. Gene networks were constructed based on published biomarkers and then enriched gene ontology terms were elucidated in subgroups using gene ontology software tools. Our results highlight the novel role of proteasome-mediated protein catabolic processes (p = 2.91 × 10−15) and plasma lipoproteins (p = 4.55 × 10−11) in anti-TNF therapy response. The results of our gene ontology analysis help elucidate the biological processes underlying non-response to anti-TNF therapy in rheumatoid arthritis and encourage further study of the highlighted processes.
Collapse
|
12
|
Metabolic Profiling in Rheumatoid Arthritis, Psoriatic Arthritis, and Psoriasis: Elucidating Pathogenesis, Improving Diagnosis, and Monitoring Disease Activity. J Pers Med 2022; 12:jpm12060924. [PMID: 35743709 PMCID: PMC9225104 DOI: 10.3390/jpm12060924] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/16/2022] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs), such as rheumatoid arthritis (RA), psoriatic arthritis (PsA), and psoriasis (Ps), represent autoinflammatory and autoimmune disorders, as well as conditions that have an overlap of both categories. Understanding the underlying pathogeneses, making diagnoses, and choosing individualized treatments remain challenging due to heterogeneous disease phenotypes and the lack of reliable biomarkers that drive the treatment choice. In this review, we provide an overview of the low-molecular-weight metabolites that might be employed as biomarkers for various applications, e.g., early diagnosis, disease activity monitoring, and treatment-response prediction, in RA, PsA, and Ps. The literature was evaluated, and putative biomarkers in different matrices were identified, categorized, and summarized. While some of these candidate biomarkers appeared to be disease-specific, others were shared across multiple IMIDs, indicating common underlying disease mechanisms. However, there is still a long way to go for their application in a routine clinical setting. We propose that studies integrating omics analyses of large patient cohorts from different IMIDs should be performed to further elucidate their pathomechanisms and treatment options. This could lead to the identification and validation of biomarkers that might be applied in the context of precision medicine to improve the clinical outcomes of these IMID patients.
Collapse
|
13
|
Yang Z, Li W, Huang H, Ren S, Men Y, Li F, Yu X, Luo Q. Detection of serum phospholipids by microchannel-integrated black phosphorus-assisted laser desorption/ionization mass spectrometry. Talanta 2022; 237:122978. [PMID: 34736700 DOI: 10.1016/j.talanta.2021.122978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 10/20/2022]
Abstract
Matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) has been widely applied in the analysis of phospholipids in biological samples. However, it remains a challenge to improve the sensitivity and reproducibility and to control the background noise of matrices. In this study, black phosphorus nanomaterial was used as the matrix of MALDI-MS, and microchannel technique was combined. This microchannel-integrated black phosphorus-assisted laser desorption/ionization (BPALDI) MS approach can effectively detect a variety of lipids with a small amount of sample, and has high sensitivity for phosphatidylcholines (PC) and lysophosphatidylcholines (LPC) with a detection limit of 0.2 μg/mL. Compared with traditional matrices, BPALDI-MS has the advantages of high sensitivity, good reproducibility, and high salt tolerance. This method was successfully applied in the detection of serum PC/LPC ratios in children patients with asthma or bronchopneumonia. This work provides a novel application of black phosphorus matrix and microchannel technique, and gives new insights into method development of rapid screening and identification of disease indicators in biological fluids.
Collapse
Affiliation(s)
- Zhiyi Yang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenbo Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hao Huang
- Shenzhen Engineering Laboratory of Single-molecule Detection and Instrument Development, Shenzhen, 518055, China
| | - Songlei Ren
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yongfan Men
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Fang Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Shenzhen Engineering Laboratory of Single-molecule Detection and Instrument Development, Shenzhen, 518055, China
| | - Xuefeng Yu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qian Luo
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Shenzhen Engineering Laboratory of Single-molecule Detection and Instrument Development, Shenzhen, 518055, China.
| |
Collapse
|
14
|
Metabolomics in Autoimmune Diseases: Focus on Rheumatoid Arthritis, Systemic Lupus Erythematous, and Multiple Sclerosis. Metabolites 2021; 11:metabo11120812. [PMID: 34940570 PMCID: PMC8708401 DOI: 10.3390/metabo11120812] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/18/2022] Open
Abstract
The metabolomics approach represents the last downstream phenotype and is widely used in clinical studies and drug discovery. In this paper, we outline recent advances in the metabolomics research of autoimmune diseases (ADs) such as rheumatoid arthritis (RA), multiple sclerosis (MuS), and systemic lupus erythematosus (SLE). The newly discovered biomarkers and the metabolic mechanism studies for these ADs are described here. In addition, studies elucidating the metabolic mechanisms underlying these ADs are presented. Metabolomics has the potential to contribute to pharmacotherapy personalization; thus, we summarize the biomarker studies performed to predict the personalization of medicine and drug response.
Collapse
|
15
|
Meehan RT, Amigues IA, Knight V. Precision Medicine for Rheumatoid Arthritis: The Right Drug for the Right Patient-Companion Diagnostics. Diagnostics (Basel) 2021; 11:diagnostics11081362. [PMID: 34441297 PMCID: PMC8391624 DOI: 10.3390/diagnostics11081362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Despite the growing number of biologic and JAK inhibitor therapeutic agents available to treat various systemic autoimmune illnesses, the lack of a validated companion diagnostic (CDx) to accurately predict drug responsiveness for an individual results in many patients being treated for years with expensive, ineffective, or toxic drugs. This review will focus primarily on rheumatoid arthritis (RA) therapeutics where the need is greatest due to poor patient outcomes if the optimum drug is delayed. We will review current FDA-approved biologic and small molecule drugs and why RA patients switch these medications. We will discuss the sampling of various tissues for potential CDx and review early results from studies investigating drug responsiveness utilizing advanced technologies including; multiplex testing of cytokines and proteins, autoantibody profiling, genomic analysis, proteomics, miRNA analysis, and metabolomics. By using these new technologies for CDx the goal is to improve RA patient outcomes and achieve similar successes like those seen in oncology using precision medicine guided therapeutics.
Collapse
Affiliation(s)
- Richard Thomas Meehan
- Department of Medicine, Rheumatology Division, National Jewish Health, Denver, CO 80206, USA;
- Correspondence:
| | - Isabelle Anne Amigues
- Department of Medicine, Rheumatology Division, National Jewish Health, Denver, CO 80206, USA;
| | - Vijaya Knight
- Immunology Department, Children’s Hospital, Aurora, CO 80045, USA;
| |
Collapse
|
16
|
Disease Differentiation and Monitoring of Anti-TNF Treatment in Rheumatoid Arthritis and Spondyloarthropathies. Int J Mol Sci 2021; 22:ijms22147389. [PMID: 34299006 PMCID: PMC8307996 DOI: 10.3390/ijms22147389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 01/16/2023] Open
Abstract
Rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) are comprehensive immunological disorders. The treatment of these disorders is limited to ameliorating the symptoms and improving the quality of life of patients. In this study, serum samples from RA, AS, and PsA patients were analyzed with metabolomic tools employing the 1H NMR method in combination with univariate and multivariate analyses. The results obtained in this study showed that the changes in metabolites were the highest for AS > RA > PsA. The study demonstrated that the time until remission or until low disease activity is achieved is shortest (approximately three months) for AS, longer for RA and longest for PsA. The statistically common metabolite that was found to be negatively correlated with the healing processes of these disorders is ethanol, which may indicate the involvement of the gut microflora and/or the breakdown of malondialdehyde as a cell membrane lipid peroxide product.
Collapse
|
17
|
Hur B, Gupta VK, Huang H, Wright KA, Warrington KJ, Taneja V, Davis JM, Sung J. Plasma metabolomic profiling in patients with rheumatoid arthritis identifies biochemical features predictive of quantitative disease activity. Arthritis Res Ther 2021; 23:164. [PMID: 34103083 PMCID: PMC8185925 DOI: 10.1186/s13075-021-02537-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic, autoimmune disorder characterized by joint inflammation and pain. In patients with RA, metabolomic approaches, i.e., high-throughput profiling of small-molecule metabolites, on plasma or serum has thus far enabled the discovery of biomarkers for clinical subgroups, risk factors, and predictors of treatment response. Despite these recent advancements, the identification of blood metabolites that reflect quantitative disease activity remains an important challenge in precision medicine for RA. Herein, we use global plasma metabolomic profiling analyses to detect metabolites associated with, and predictive of, quantitative disease activity in patients with RA. Methods Ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was performed on a discovery cohort consisting of 128 plasma samples from 64 RA patients and on a validation cohort of 12 samples from 12 patients. The resulting metabolomic profiles were analyzed with two different strategies to find metabolites associated with RA disease activity defined by the Disease Activity Score-28 using C-reactive protein (DAS28-CRP). More specifically, mixed-effects regression models were used to identify metabolites differentially abundant between two disease activity groups (“lower”, DAS28-CRP ≤ 3.2; and “higher”, DAS28-CRP > 3.2) and to identify metabolites significantly associated with DAS28-CRP scores. A generalized linear model (GLM) was then constructed for estimating DAS28-CRP using plasma metabolite abundances. Finally, for associating metabolites with CRP (an indicator of inflammation), metabolites differentially abundant between two patient groups (“low-CRP”, CRP ≤ 3.0 mg/L; “high-CRP”, CRP > 3.0 mg/L) were investigated. Results We identified 33 metabolites differentially abundant between the lower and higher disease activity groups (P < 0.05). Additionally, we identified 51 metabolites associated with DAS28-CRP (P < 0.05). A GLM based upon these 51 metabolites resulted in higher prediction accuracy (mean absolute error [MAE] ± SD: 1.51 ± 1.77) compared to a GLM without feature selection (MAE ± SD: 2.02 ± 2.21). The predictive value of this feature set was further demonstrated on a validation cohort of twelve plasma samples, wherein we observed a stronger correlation between predicted and actual DAS28-CRP (with feature selection: Spearman’s ρ = 0.69, 95% CI: [0.18, 0.90]; without feature selection: Spearman’s ρ = 0.18, 95% CI: [−0.44, 0.68]). Lastly, among all identified metabolites, the abundances of eight were significantly associated with the CRP patient groups while controlling for potential confounders (P < 0.05). Conclusions We demonstrate for the first time the prediction of quantitative disease activity in RA using plasma metabolomes. The metabolites identified herein provide insight into circulating pro-/anti-inflammatory metabolic signatures that reflect disease activity and inflammatory status in RA patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02537-4.
Collapse
Affiliation(s)
- Benjamin Hur
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Vinod K Gupta
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Harvey Huang
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, USA
| | - Kerry A Wright
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kenneth J Warrington
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Veena Taneja
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - John M Davis
- Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jaeyun Sung
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA. .,Division of Surgery Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA. .,Division of Rheumatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Ishibashi Y, Harada S, Takeuchi A, Iida M, Kurihara A, Kato S, Kuwabara K, Hirata A, Shibuki T, Okamura T, Sugiyama D, Sato A, Amano K, Hirayama A, Sugimoto M, Soga T, Tomita M, Takebayashi T. Reliability of urinary charged metabolite concentrations in a large-scale cohort study using capillary electrophoresis-mass spectrometry. Sci Rep 2021; 11:7407. [PMID: 33795760 PMCID: PMC8016858 DOI: 10.1038/s41598-021-86600-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Currently, large-scale cohort studies for metabolome analysis have been launched globally. However, only a few studies have evaluated the reliability of urinary metabolome analysis. This study aimed to establish the reliability of urinary metabolomic profiling in cohort studies. In the Tsuruoka Metabolomics Cohort Study, 123 charged metabolites were identified and routinely quantified using capillary electrophoresis-mass spectrometry (CE-MS). We evaluated approximately 750 quality control (QC) samples and 6,720 participants’ spot urine samples. We calculated inter- and intra-batch coefficients of variation in the QC and participant samples and technical intraclass correlation coefficients (ICC). A correlation of metabolite concentrations between spot and 24-h urine samples obtained from 32 sub-cohort participants was also evaluated. The coefficient of variation (CV) was less than 20% for 87 metabolites (70.7%) and 20–30% for 19 metabolites (15.4%) in the QC samples. There was less than 20% inter-batch CV for 106 metabolites (86.2%). Most urinary metabolites would have reliability for measurement. The 96 metabolites (78.0%) was above 0.75 for the estimated ICC, and those might be useful for epidemiological analysis. Among individuals, the Pearson correlation coefficient of 24-h and spot urine was more than 70% for 59 of the 99 metabolites. These results show that the profiling of charged metabolites using CE-MS in morning spot human urine is suitable for epidemiological metabolomics studies.
Collapse
Affiliation(s)
- Yoshiki Ishibashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan.,Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Ayano Takeuchi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Miho Iida
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Ayako Kurihara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Suzuka Kato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Kazuyo Kuwabara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Aya Hirata
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Takuma Shibuki
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Tomonori Okamura
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | - Daisuke Sugiyama
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan.,Faculty of Nursing And Medical Care, Keio University, Fujisawa, Kanagawa, Japan
| | - Asako Sato
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Kaori Amano
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.,Faculty of Environment and Information Studies, Keio University, Fujisawa, Kanagawa, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.,Faculty of Environment and Information Studies, Keio University, Fujisawa, Kanagawa, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan. .,Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.
| |
Collapse
|
19
|
Prediction of response of methotrexate in patients with rheumatoid arthritis using serum lipidomics. Sci Rep 2021; 11:7266. [PMID: 33790392 PMCID: PMC8012618 DOI: 10.1038/s41598-021-86729-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/15/2021] [Indexed: 01/31/2023] Open
Abstract
Methotrexate (MTX) is a common first-line treatment for new-onset rheumatoid arthritis (RA). However, MTX is ineffective for 30–40% of patients and there is no way to know which patients might benefit. Here, we built statistical models based on serum lipid levels measured at two time-points (pre-treatment and following 4 weeks on-drug) to investigate if MTX response (by 6 months) could be predicted. Patients about to commence MTX treatment for the first time were selected from the Rheumatoid Arthritis Medication Study (RAMS). Patients were categorised as good or non-responders following 6 months on-drug using EULAR response criteria. Serum lipids were measured using ultra‐performance liquid chromatography–mass spectrometry and supervised machine learning methods (including regularized regression, support vector machine and random forest) were used to predict EULAR response. Models including lipid levels were compared to models including clinical covariates alone. The best performing classifier including lipid levels (assessed at 4 weeks) was constructed using regularized regression (ROC AUC 0.61 ± 0.02). However, the clinical covariate based model outperformed the classifier including lipid levels when either pre- or on-treatment time-points were investigated (ROC AUC 0.68 ± 0.02). Pre- or early-treatment serum lipid profiles are unlikely to inform classification of MTX response by 6 months with performance adequate for use in RA clinical management.
Collapse
|
20
|
Jutley GS, Sahota K, Sahbudin I, Filer A, Arayssi T, Young SP, Raza K. Relationship Between Inflammation and Metabolism in Patients With Newly Presenting Rheumatoid Arthritis. Front Immunol 2021; 12:676105. [PMID: 34650548 PMCID: PMC8507469 DOI: 10.3389/fimmu.2021.676105] [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: 03/04/2021] [Accepted: 08/04/2021] [Indexed: 12/29/2022] Open
Abstract
Background Systemic inflammation in rheumatoid arthritis (RA) is associated with metabolic changes. We used nuclear magnetic resonance (NMR) spectroscopy-based metabolomics to assess the relationship between an objective measure of systemic inflammation [C-reactive protein (CRP)] and both the serum and urinary metabolome in patients with newly presenting RA. Methods Serum (n=126) and urine (n=83) samples were collected at initial presentation from disease modifying anti-rheumatic drug naïve RA patients for metabolomic profile assessment using 1-dimensional 1H-NMR spectroscopy. Metabolomics data were analysed using partial least square regression (PLS-R) and orthogonal projections to latent structure discriminant analysis (OPLS-DA) with cross validation. Results Using PLS-R analysis, a relationship between the level of inflammation, as assessed by CRP, and the serum (p=0.001) and urinary (p<0.001) metabolome was detectable. Likewise, following categorisation of CRP into tertiles, patients in the lowest CRP tertile and the highest CRP tertile were statistically discriminated using OPLS-DA analysis of both serum (p=0.033) and urinary (p<0.001) metabolome. The most highly weighted metabolites for these models included glucose, amino acids, lactate, and citrate. These findings suggest increased glycolysis, perturbation in the citrate cycle, oxidative stress, protein catabolism and increased urea cycle activity are key characteristics of newly presenting RA patients with elevated CRP. Conclusions This study consolidates our understanding of a previously identified relationship between serum metabolite profile and inflammation and provides novel evidence that there is a relationship between urinary metabolite profile and inflammation as measured by CRP. Identification of these metabolic perturbations provides insights into the pathogenesis of RA and may help in the identification of therapeutic targets.
Collapse
Affiliation(s)
- Gurpreet Singh Jutley
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Kalvin Sahota
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Ilfita Sahbudin
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Andrew Filer
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Research Into Inflammatory Arthritis Centre, Versus Arthritis, University of Birmingham, Birmingham, United Kingdom
| | | | - Stephen P Young
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Karim Raza
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and Institute for Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Research Into Inflammatory Arthritis Centre, Versus Arthritis, University of Birmingham, Birmingham, United Kingdom.,Department of Rheumatology, Sandwell and West Birmingham NHS Trust, Birmingham, United Kingdom
| |
Collapse
|
21
|
Kishikawa T, Maeda Y, Nii T, Arase N, Hirata J, Suzuki K, Yamamoto K, Masuda T, Ogawa K, Tsuji S, Matsushita M, Matsuoka H, Yoshimura M, Tsunoda S, Ohshima S, Narazaki M, Ogata A, Saeki Y, Inohara H, Kumanogoh A, Takeda K, Okada Y. Increased levels of plasma nucleotides in patients with rheumatoid arthritis. Int Immunol 2020; 33:119-124. [PMID: 32866240 PMCID: PMC7846180 DOI: 10.1093/intimm/dxaa059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/26/2020] [Indexed: 01/04/2023] Open
Abstract
Novel biomarkers of rheumatoid arthritis (RA), in addition to antibodies against cyclic citrullinated peptides, are required. Metabolome analysis is a promising approach to identify metabolite biomarkers for clinical diagnosis. We adopted a comprehensive non-targeted metabolomics approach combining capillary electrophoresis time-of-flight mass spectrometry (TOFMS) and liquid chromatography TOFMS. We constructed metabolomics profiling of 286 plasma samples of a Japanese population [92 RA patients, 13 systemic lupus erythematosus (SLE) patients and 181 healthy controls). RA case–control association tests showed that seven metabolites exhibited significantly increased levels in RA samples compared with controls (P < 1.0 × 10−4; UTP, ethanolamine phosphate, ATP, GDP, ADP, 6-aminohexanoic acid and taurine), whereas one exhibited a decreased level (xanthine). The plasma levels of these eight metabolites were not significantly different between seropositive and seronegative RA patients (P > 0.05; n = 68 and 24, respectively). The four nucleotide levels (UTP, ATP, GDP and ADP) were significantly higher in the non-treatment patients in comparison between patients with and without treatment (P < 0.014; n = 57 and 35, respectively). Furthermore, we found that none of the four nucleotide levels showed significant differences in SLE case–control association tests (P > 0.2; 13 patients with SLE and the 181 shared controls) and psoriatic arthritis (PsA) case–control association tests (P > 0.11; 42 patients with PsA and 38 healthy controls), indicating disease specificity in RA. In conclusion, our large-scale metabolome analysis demonstrated the increased plasma nucleotide levels in RA patients, which could be used as potential clinical biomarkers of RA, especially for seronegative RA.
Collapse
Affiliation(s)
- Toshihiro Kishikawa
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichi Maeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan.,Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takuro Nii
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan.,Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Noriko Arase
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jun Hirata
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ken Suzuki
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tatsuo Masuda
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kotaro Ogawa
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shigeyoshi Tsuji
- Department of Orthopedics/Rheumatology, NHO Osaka Minami Medical Center, Kawachinagano, Japan
| | - Masato Matsushita
- Department of Rheumatology and Allergology, Saiseikai Senri Hospital, Suita, Japan.,Rheumatology and Allergology, NHO Osaka Minami Medical Center, Kawachinagano, Japan
| | - Hidetoshi Matsuoka
- Rheumatology and Allergology, NHO Osaka Minami Medical Center, Kawachinagano, Japan
| | - Maiko Yoshimura
- Rheumatology and Allergology, NHO Osaka Minami Medical Center, Kawachinagano, Japan
| | | | - Shiro Ohshima
- Rheumatology and Allergology, NHO Osaka Minami Medical Center, Kawachinagano, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsushi Ogata
- Division of Rheumatology, Department of Internal Medicine, Daini Osaka Police Hospital, Osaka, Japan
| | - Yukihiko Saeki
- Rheumatology and Allergology, NHO Osaka Minami Medical Center, Kawachinagano, Japan.,Clinical Research, NHO Osaka Minami Medical Center, Kawachinagano, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
22
|
Daly R, Blackburn G, Best C, Goodyear CS, Mudaliar M, Burgess K, Stirling A, Porter D, McInnes IB, Barrett MP, Dale J. Changes in Plasma Itaconate Elevation in Early Rheumatoid Arthritis Patients Elucidates Disease Activity Associated Macrophage Activation. Metabolites 2020; 10:metabo10060241. [PMID: 32531990 PMCID: PMC7344783 DOI: 10.3390/metabo10060241] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/05/2020] [Accepted: 06/07/2020] [Indexed: 12/29/2022] Open
Abstract
Changes in the plasma metabolic profile were characterised in newly diagnosed rheumatoid arthritis (RA) patients upon commencement of conventional disease-modifying anti-rheumatic drug (cDMARD) therapy. Plasma samples collected in an early RA randomised strategy study (NCT00920478) that compared clinical (DAS) disease activity assessment with musculoskeletal ultrasound assessment (MSUS) to drive treatment decisions were subjected to untargeted metabolomic analysis. Metabolic profiles were collected at pre- and three months post-commencement of nonbiologic cDMARD. Metabolites that changed in association with changes in the DAS44 score were identified at the three-month timepoint. A total of nine metabolites exhibited a clear correlation with a reduction in DAS44 score following cDMARD commencement, particularly itaconate, its derived anhydride and a derivative of itaconate CoA. Increasing itaconate correlated with improved DAS44 score and decreasing levels of C-reactive protein (CRP). cDMARD treatment effects invoke consistent changes in plasma detectable metabolites, that in turn implicate clinical disease activity with macrophages. Such changes inform RA pathogenesis and reveal for the first time a link between itaconate production and resolution of inflammatory disease in humans. Quantitative metabolic biomarker-based tests of clinical change in state are feasible and should be developed around the itaconate pathway.
Collapse
Affiliation(s)
- Rónán Daly
- Glasgow Polyomics, University of Glasgow, Glasgow G61 1BD, UK; (R.D.); (G.B.); (M.M.); (K.B.); (M.P.B.)
| | - Gavin Blackburn
- Glasgow Polyomics, University of Glasgow, Glasgow G61 1BD, UK; (R.D.); (G.B.); (M.M.); (K.B.); (M.P.B.)
| | - Cameron Best
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
| | - Carl S. Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
| | - Manikhandan Mudaliar
- Glasgow Polyomics, University of Glasgow, Glasgow G61 1BD, UK; (R.D.); (G.B.); (M.M.); (K.B.); (M.P.B.)
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
- Institute of Biodiversity Animal Health and Comparative Medicine, University of Glasgow, Bearsden Road, Glasgow G61 1QH, UK
| | - Karl Burgess
- Glasgow Polyomics, University of Glasgow, Glasgow G61 1BD, UK; (R.D.); (G.B.); (M.M.); (K.B.); (M.P.B.)
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
- Institute of Quantitative Biology, Biochemistry and Biotechnology, The University of Edinburgh, Edinburgh EH9 3FF, UK
| | - Anne Stirling
- Department of Rheumatology, Gartnavel General Hospital, Glasgow G12 0YN, UK;
| | - Duncan Porter
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
- Department of Rheumatology, Gartnavel General Hospital, Glasgow G12 0YN, UK;
| | - Iain B. McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
| | - Michael P. Barrett
- Glasgow Polyomics, University of Glasgow, Glasgow G61 1BD, UK; (R.D.); (G.B.); (M.M.); (K.B.); (M.P.B.)
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
| | - James Dale
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK; (C.B.); (C.S.G.); (D.P.); (I.B.M.)
- Department of Rheumatology, Wishaw General Hospital, 50 Netherton Street, Wishaw, North Lanarkshire ML2 0DP, UK
- Correspondence:
| |
Collapse
|
23
|
Takahashi S, Saegusa J, Onishi A, Morinobu A. Biomarkers identified by serum metabolomic analysis to predict biologic treatment response in rheumatoid arthritis patients. Rheumatology (Oxford) 2020; 58:2153-2161. [PMID: 31143951 DOI: 10.1093/rheumatology/kez199] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/23/2019] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Biologic treatment has recently revolutionized the management of RA. Despite this success, ∼30-40% of the patients undergoing biologic treatment respond insufficiently. The aim of this study was to identify several specific reliable metabolites for predicting the response of RA patients to TNF-α inhibitors (TNFi) and abatacept (ABT), using capillary electrophoresis-time-of-flight mass spectrometry (CE-TOFMS). METHODS We collected serum from RA patients with moderate or high disease activity prior to biologic treatment, and obtained the serum metabolomic profiles of these samples using CE-TOFMS. The patients' response was determined 12 weeks after starting biologic treatment, according to the EULAR response criteria. We compared the metabolites between the response and non-response patient groups and analysed their discriminative ability. RESULTS Among 43 total patients, 14 of 26 patients in the TNFi group and 6 of 17 patients in the ABT group responded to the biologic treatment. Of the metabolites separated by CE-TOFMS, 196 were identified as known substances. Using an orthogonal partial least-squares discriminant analysis, we identified five metabolites as potential predictors of TNFi responders and three as predictors of ABT responders. Receiver operating characteristic analyses for multiple biomarkers revealed an area under the curve (AUC) of 0.941, with a sensitivity of 85.7% and specificity of 100% for TNFi, and an AUC of 0.985, with a sensitivity of 100% and specificity of 90.9% for ABT. CONCLUSION By metabolomic analysis, we identified serum biomarkers that have a high ability to predict the response of RA patients to TNFi or ABT treatment.
Collapse
Affiliation(s)
- Soshi Takahashi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe,Japan.,Centre for Rheumatic Disease, Shinko Hospital, Kobe,Japan
| | - Jun Saegusa
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe,Japan
| | - Akira Onishi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe,Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Kobe,Japan
| |
Collapse
|
24
|
Funk RS, Singh RK, Becker ML. Metabolomic Profiling to Identify Molecular Biomarkers of Cellular Response to Methotrexate In Vitro. Clin Transl Sci 2020; 13:137-146. [PMID: 31651077 PMCID: PMC6951846 DOI: 10.1111/cts.12694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/06/2019] [Indexed: 01/21/2023] Open
Abstract
Variation in methotrexate (MTX) efficacy represents a significant barrier to early and effective disease control in the treatment of autoimmune arthritis. We hypothesize that the utilization of metabolomic techniques will allow for an improved understanding of the biochemical basis for the pharmacological activity of MTX, and can promote the identification and evaluation of novel molecular biomarkers of MTX response. In this work, erythroblastoid cells were exposed to MTX at the physiologic concentration of 1,000 nM and analyzed using three metabolomic platforms to give a broad spectrum of cellular metabolites. MTX pharmacological activity, defined as cellular growth inhibition, was associated with an altered cellular metabolomic profile based on the analysis of 724 identified metabolites. By discriminant analysis, MTX treatment was associated with increases in ketoisovaleric acid, fructose, galactose, and 2-deoxycytidine, and corresponding reductions in 2-deoxyuridine, phosphatidylinositol 32:0, orotic acid, and inosine monophosphate. Inclusion of data from analysis of folate metabolism in combination with chemometric and metabolic network analysis demonstrated that MTX treatment is associated with dysregulated folate metabolism and nucleotide biosynthesis, which is in line with its known mechanism of action. However, MTX treatment was also associated with alterations in a diversity of metabolites, including intermediates of amino acid, carbohydrate, and lipid metabolism. Collectively, these findings support a robust metabolic response following exposure to physiologic concentrations of MTX. They also identify various metabolic intermediates that are associated with the pharmacological activity of MTX, and are, therefore, potential molecular biomarker candidates in future preclinical and clinical studies of MTX efficacy in autoimmune arthritis.
Collapse
Affiliation(s)
- Ryan S. Funk
- Department of Pharmacy PracticeMedical CenterUniversity of KansasKansas CityKansasUSA
| | - Rakesh K. Singh
- Department of Pharmacy PracticeMedical CenterUniversity of KansasKansas CityKansasUSA
| | - Mara L. Becker
- Division of RheumatologyDepartment of PediatricsDuke Children's HospitalDurhamNorth CarolinaUSA
| |
Collapse
|
25
|
Gladine C, Ostermann AI, Newman JW, Schebb NH. MS-based targeted metabolomics of eicosanoids and other oxylipins: Analytical and inter-individual variabilities. Free Radic Biol Med 2019; 144:72-89. [PMID: 31085232 DOI: 10.1016/j.freeradbiomed.2019.05.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/19/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
Oxylipins, including the well-known eicosanoids, are potent lipid mediators involved in numerous physiological and pathological processes. Therefore, their quantitative profiling has gained a lot of attention during the last years notably in the active field of health biomarker discovery. Oxylipins include hundreds of structurally and stereochemically distinct lipid species which today are most commonly analyzed by (ultra) high performance liquid chromatography-mass spectrometry based ((U)HPLC-MS) methods. To maximize the utility of oxylipin profiling in clinical research, it is crucial to understand and assess the factors contributing to the analytical and biological variability of oxylipin profiles in humans. In this review, these factors and their impacts are summarized and discussed, providing a framework for recommendations expected to enhance the interlaboratory comparability and biological interpretation of oxylipin profiling in clinical research.
Collapse
Affiliation(s)
- Cécile Gladine
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France.
| | - Annika I Ostermann
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, Gaußstraße 20, University of Wuppertal, 42119, Wuppertal, Germany
| | - John W Newman
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; University of California Davis, Department of Nutrition, Davis, CA, USA
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, Gaußstraße 20, University of Wuppertal, 42119, Wuppertal, Germany
| |
Collapse
|
26
|
Kim J, Kang SC, Yoon NE, Kim Y, Choi J, Park N, Jung H, Jung BH, Ju JH. Metabolomic profiles of induced pluripotent stem cells derived from patients with rheumatoid arthritis and osteoarthritis. Stem Cell Res Ther 2019; 10:319. [PMID: 31730022 PMCID: PMC6858676 DOI: 10.1186/s13287-019-1408-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/17/2019] [Accepted: 09/03/2019] [Indexed: 12/19/2022] Open
Abstract
Background Metabolomics is the systemic study of the unique fingerprints of metabolites involved in cellular processes and biochemical reactions. The metabolomic approach is useful in diagnosing and predicting the development of rheumatoid arthritis (RA) and osteoarthritis (OA) and is emerging as a useful tool for identifying disease biomarkers. The aim of this study was to compare the metabolic blueprint of fibroblast-like synoviocyte (FLS) cells and induced pluripotent stem cells (iPSCs) derived from RA and OA patients. Methods Somatic cells of RA patients (n = 3) and OA patients (n = 3) were isolated, transduced with a lentiviral plasmid, and reprogrammed into iPSCs displaying pluripotency. Metabolic profiling of RA and OA patient–derived FLS cells and iPSCs was performed using liquid chromatography/mass spectrometry and statistical analysis. After normalization by the sum of the peak intensities through LC/MS, 37 metabolites were detected across RA and OA patients. Results The metabolites of RA and OA were distinguishable according to the PLS-DA analysis. LysoPC (20:4), 4-methoxychalcone, phosphorylcholine, and nicotinamide (NAM) were significantly higher in RA iPSCs than in OA iPSCs (p < 0.05). The NMNAT-3 enzyme, which catalyzes an important step in the biosynthesis of NAD+ from adenosine triphosphate, was also upregulated in RA iPSCs. Interestingly, the proliferation of RA iPSCs was significantly greater than OA iPSC proliferation (p < 0.05). NAM played a critical role in the proliferation of RA iPSCs but not in OA iPSCs. When iPSCs were treated with 100 nM of the NAM inhibitor tannic acid (TA), the proliferation of RA iPSCs was significantly reduced (p < 0.001). Conclusions The metabolites of RA and OA FLS cells and RA and OA iPSCs were all clearly distinguishable from each other. NAM played a critical role in the proliferation of RA iPSCs but not in OA iPSCs. TA effectively inhibited the expression of NAM in RA iPSCs and is a possible effective treatment for RA patients.
Collapse
Affiliation(s)
- Juryun Kim
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea
| | | | - Na Eun Yoon
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Yena Kim
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea
| | - Jinhyeok Choi
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea
| | - Narae Park
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea
| | - Hyerin Jung
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea
| | - Byung Hwa Jung
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea. .,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 137-701, South Korea. .,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
27
|
Sasaki C, Hiraishi T, Oku T, Okuma K, Suzumura K, Hashimoto M, Ito H, Aramori I, Hirayama Y. Metabolomic approach to the exploration of biomarkers associated with disease activity in rheumatoid arthritis. PLoS One 2019; 14:e0219400. [PMID: 31295280 PMCID: PMC6622493 DOI: 10.1371/journal.pone.0219400] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/21/2019] [Indexed: 11/19/2022] Open
Abstract
We aimed to investigate metabolites associated with the 28-joint disease activity score based on erythrocyte sedimentation rate (DAS28-ESR) in patients with rheumatoid arthritis (RA) using capillary electrophoresis quadrupole time-of-flight mass spectrometry. Plasma and urine samples were collected from 32 patients with active RA (DAS28-ESR≥3.2) and 17 with inactive RA (DAS28-ESR<3.2). We found 15 metabolites in plasma and 20 metabolites in urine which showed a significant but weak positive or negative correlation with DAS28-ESR. When metabolites between active and inactive patients were compared, 9 metabolites in plasma and 15 in urine were found to be significantly different. Consequently, we selected 11 metabolites in plasma and urine as biomarker candidates which significantly correlated positively or negatively with DAS28-ESR, and significantly differed between active and inactive patients. When a multiple logistic regression model was built to discriminate active and inactive cohorts, three variables-histidine and guanidoacetic acid from plasma and hypotaurine from urine-generated a high area under the receiver operating characteristic (ROC) curve value (AUC = 0.8934). Thus, this metabolomics approach appeared to be useful for investigating biomarkers of RA. Combination of plasma and urine analysis may lead to more precise and reliable understanding of the disease condition. We also considered the pathophysiological significance of the found biomarker candidates.
Collapse
Affiliation(s)
- Chiyomi Sasaki
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Tomoko Hiraishi
- Analysis & Pharmacokinetics Research Labs., Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Takuma Oku
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Kenji Okuma
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Kenichi Suzumura
- Analysis & Pharmacokinetics Research Labs., Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Hiromu Ito
- Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
| | - Ichiro Aramori
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
| | - Yoshitaka Hirayama
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
28
|
Can Metabolic Pathways Be Therapeutic Targets in Rheumatoid Arthritis? J Clin Med 2019; 8:jcm8050753. [PMID: 31137815 PMCID: PMC6572063 DOI: 10.3390/jcm8050753] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/05/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
The metabolic rewiring of tumor cells and immune cells has been viewed as a promising source of novel drug targets. Many of the molecular pathways implicated in rheumatoid arthritis (RA) directly modify synovium metabolism and transform the resident cells, such as the fibroblast-like synoviocytes (FLS), and the synovial tissue macrophages (STM), toward an overproduction of enzymes, which degrade cartilage and bone, and cytokines, which promote immune cell infiltration. Recent studies have shown metabolic changes in stromal and immune cells from RA patients. Metabolic disruption in the synovium provide the opportunity to use in vivo metabolism-based imaging techniques for patient stratification and to monitor treatment response. In addition, these metabolic changes may be therapeutically targetable. Thus, resetting metabolism of the synovial membrane offers additional opportunities for disease modulation and restoration of homeostasis in RA. In fact, rheumatologists already use the antimetabolite methotrexate, a chemotherapy agent, for the treatment of patients with inflammatory arthritis. Metabolic targets that do not compromise systemic homeostasis or corresponding metabolic functions in normal cells could increase the drug armamentarium in rheumatic diseases for combination therapy independent of systemic immunosuppression. This article summarizes what is known about metabolism in synovial tissue cells and highlights chemotherapies that target metabolism as potential future therapeutic strategies for RA.
Collapse
|
29
|
Archer R, Hock E, Hamilton J, Stevens J, Essat M, Poku E, Clowes M, Pandor A, Stevenson M. Assessing prognosis and prediction of treatment response in early rheumatoid arthritis: systematic reviews. Health Technol Assess 2019; 22:1-294. [PMID: 30501821 DOI: 10.3310/hta22660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic, debilitating disease associated with reduced quality of life and substantial costs. It is unclear which tests and assessment tools allow the best assessment of prognosis in people with early RA and whether or not variables predict the response of patients to different drug treatments. OBJECTIVE To systematically review evidence on the use of selected tests and assessment tools in patients with early RA (1) in the evaluation of a prognosis (review 1) and (2) as predictive markers of treatment response (review 2). DATA SOURCES Electronic databases (e.g. MEDLINE, EMBASE, The Cochrane Library, Web of Science Conference Proceedings; searched to September 2016), registers, key websites, hand-searching of reference lists of included studies and key systematic reviews and contact with experts. STUDY SELECTION Review 1 - primary studies on the development, external validation and impact of clinical prediction models for selected outcomes in adult early RA patients. Review 2 - primary studies on the interaction between selected baseline covariates and treatment (conventional and biological disease-modifying antirheumatic drugs) on salient outcomes in adult early RA patients. RESULTS Review 1 - 22 model development studies and one combined model development/external validation study reporting 39 clinical prediction models were included. Five external validation studies evaluating eight clinical prediction models for radiographic joint damage were also included. c-statistics from internal validation ranged from 0.63 to 0.87 for radiographic progression (different definitions, six studies) and 0.78 to 0.82 for the Health Assessment Questionnaire (HAQ). Predictive performance in external validations varied considerably. Three models [(1) Active controlled Study of Patients receiving Infliximab for the treatment of Rheumatoid arthritis of Early onset (ASPIRE) C-reactive protein (ASPIRE CRP), (2) ASPIRE erythrocyte sedimentation rate (ASPIRE ESR) and (3) Behandelings Strategie (BeSt)] were externally validated using the same outcome definition in more than one population. Results of the random-effects meta-analysis suggested substantial uncertainty in the expected predictive performance of models in a new sample of patients. Review 2 - 12 studies were identified. Covariates examined included anti-citrullinated protein/peptide anti-body (ACPA) status, smoking status, erosions, rheumatoid factor status, C-reactive protein level, erythrocyte sedimentation rate, swollen joint count (SJC), body mass index and vascularity of synovium on power Doppler ultrasound (PDUS). Outcomes examined included erosions/radiographic progression, disease activity, physical function and Disease Activity Score-28 remission. There was statistical evidence to suggest that ACPA status, SJC and PDUS status at baseline may be treatment effect modifiers, but not necessarily that they are prognostic of response for all treatments. Most of the results were subject to considerable uncertainty and were not statistically significant. LIMITATIONS The meta-analysis in review 1 was limited by the availability of only a small number of external validation studies. Studies rarely investigated the interaction between predictors and treatment. SUGGESTED RESEARCH PRIORITIES Collaborative research (including the use of individual participant data) is needed to further develop and externally validate the clinical prediction models. The clinical prediction models should be validated with respect to individual treatments. Future assessments of treatment by covariate interactions should follow good statistical practice. CONCLUSIONS Review 1 - uncertainty remains over the optimal prediction model(s) for use in clinical practice. Review 2 - in general, there was insufficient evidence that the effect of treatment depended on baseline characteristics. STUDY REGISTRATION This study is registered as PROSPERO CRD42016042402. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Rachel Archer
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Emma Hock
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Jean Hamilton
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - John Stevens
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Munira Essat
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Edith Poku
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Mark Clowes
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Abdullah Pandor
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Matt Stevenson
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| |
Collapse
|
30
|
Lequerré T, Rottenberg P, Derambure C, Cosette P, Vittecoq O. Predictors of treatment response in rheumatoid arthritis. Joint Bone Spine 2019; 86:151-158. [DOI: 10.1016/j.jbspin.2018.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 12/13/2022]
|
31
|
Gupta L, Ahmed S, Jain A, Misra R. Emerging role of metabolomics in rheumatology. Int J Rheum Dis 2018; 21:1468-1477. [PMID: 30146741 DOI: 10.1111/1756-185x.13353] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/21/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022]
Abstract
The pursuit for understanding disease pathogenesis, in this age of rapid laboratory diagnostics and fast-paced research, has led scientists worldwide to take recourse in hypothesis-free approaches for molecular diagnosis. Metabolomics is one such powerful tool that explores comprehensibly the metabolic alternations in human diseases. It involves study of small molecules of less than 1 kD in size by either LSMS or nuclear magnetic resonance. Unlike genomics, which tells us what may have happened, metabolomics reflects what did happen. The NMR technique has an advantage of analyzing metabolites without sample preparation, thereby diminishing artifacts, is less cumbersome and with the latest database on Metabolome; about 30 000 metabolites can be identified. The study of metabolomics for several rheumatic diseases, including rheumatoid arthritis, lupus, osteoarthritis and vasculitis, has revealed distinctive metabolic signatures. Thus, metabolomics is a technique that promises precision medicine with better biomarkers, robust predictors of drug response and of disease outcome, discovery of newer metabolites and pathways in disease pathogenesis, and finally, targeted drug development. This review intends to decipher its relevance in common rheumatic diseases.
Collapse
Affiliation(s)
- Latika Gupta
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Sakir Ahmed
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Avinash Jain
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Ramnath Misra
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
32
|
Teitsma XM, Yang W, Jacobs JWG, Pethö-Schramm A, Borm MEA, Harms AC, Hankemeier T, van Laar JM, Bijlsma JWJ, Lafeber FPJG. Baseline metabolic profiles of early rheumatoid arthritis patients achieving sustained drug-free remission after initiating treat-to-target tocilizumab, methotrexate, or the combination: insights from systems biology. Arthritis Res Ther 2018; 20:230. [PMID: 30322408 PMCID: PMC6235217 DOI: 10.1186/s13075-018-1729-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/20/2018] [Indexed: 12/26/2022] Open
Abstract
Background We previously identified, in newly diagnosed rheumatoid arthritis (RA) patients, networks of co-expressed genes and proteomic biomarkers associated with achieving sustained drug-free remission (sDFR) after treatment with tocilizumab- or methotrexate-based strategies. The aim of this study was to identify, within the same patients, metabolic pathways important for achieving sDFR and to subsequently study the complex interactions between different components of the biological system and how these interactions might affect the therapeutic response in early RA. Methods Serum samples were analyzed of 60 patients who participated in the U-Act-Early trial (ClinicalTrials.gov number NCT01034137) and initiated treatment with methotrexate, tocilizumab, or the combination and who were thereafter able to achieve sDFR (n = 37); as controls, patients were selected who never achieved a drug-free status (n = 23). Metabolomic measurements were performed using mass spectrometry on oxidative stress, amine, and oxylipin platforms covering various compounds. Partial least square discriminant analyses (PLSDA) were performed to identify, per strategy arm, relevant metabolites of which the biological pathways were studied. In addition, integrative analyses were performed correlating the previously identified transcripts and proteins with the relevant metabolites. Results In the tocilizumab plus methotrexate, tocilizumab, and methotrexate strategy, respectively, 19, 13, and 12 relevant metabolites were found, which were subsequently used for pathway analyses. The most significant pathway in the tocilizumab plus methotrexate strategy was “histidine metabolism” (p < 0.001); in the tocilizumab strategy it was “arachidonic acid metabolism” (p = 0.018); and in the methotrexate strategy it was “arginine and proline metabolism” (p = 0.022). These pathways have treatment-specific drug interactions with metabolites affecting either the signaling of interleukin-6, which is inhibited by tocilizumab, or affecting protein synthesis from amino acids, which is inhibited by methotrexate. Conclusion In early RA patients treated-to-target with a tocilizumab- or methotrexate-based strategy, several metabolites were found to be associated with achieving sDFR. In line with our previous observations, by analyzing relevant transcripts and proteins within the same patients, the metabolic profiles were found to be different between the strategy arms. Our metabolic analysis further supports the hypothesis that achieving sDFR is not only dependent on predisposing biomarkers, but also on the specific treatment that has been initiated. Trial registration ClinicalTrials.gov, NCT01034137. Registered on January 2010 Electronic supplementary material The online version of this article (10.1186/s13075-018-1729-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xavier M Teitsma
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands.
| | - Wei Yang
- Leiden Academic Center for Drug Research, Leiden University, 2300 RA, Leiden, Netherlands
| | - Johannes W G Jacobs
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands
| | | | | | - Amy C Harms
- Leiden Academic Center for Drug Research, Leiden University, 2300 RA, Leiden, Netherlands.,Netherlands Metabolomic Centre, Einsteinweg 55, 2333 CC, Leiden, Netherlands
| | - Thomas Hankemeier
- Leiden Academic Center for Drug Research, Leiden University, 2300 RA, Leiden, Netherlands.,Netherlands Metabolomic Centre, Einsteinweg 55, 2333 CC, Leiden, Netherlands
| | - Jacob M van Laar
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands
| | - Johannes W J Bijlsma
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands
| | - Floris P J G Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands
| |
Collapse
|
33
|
Anderson JR, Chokesuwattanaskul S, Phelan MM, Welting TJM, Lian LY, Peffers MJ, Wright HL. 1H NMR Metabolomics Identifies Underlying Inflammatory Pathology in Osteoarthritis and Rheumatoid Arthritis Synovial Joints. J Proteome Res 2018; 17:3780-3790. [PMID: 30229649 PMCID: PMC6220363 DOI: 10.1021/acs.jproteome.8b00455] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Despite
osteoarthritis (OA) and rheumatoid arthritis (RA) being typically
age-related, their underlying etiologies are markedly different. We
used 1H nuclear magnetic resonance (NMR) spectroscopy to
identify differences in metabolite profiles in low volumes of OA and
RA synovial fluid (SF). SF was aspirated from knee joints of 10 OA
and 14 RA patients. 100 μL SF was analyzed using a 700 MHz Avance
IIIHD Bruker NMR spectrometer with a TCI cryoprobe. Spectra were analyzed
by Chenomx, Bruker TopSpin and AMIX software. Statistical analysis
was undertaken using Metaboanalyst. 50 metabolites were annotated,
including amino acids, saccharides, nucleotides and soluble lipids.
Discriminant analysis identified group separation between OA and RA
cohorts, with 32 metabolites significantly different between OA and
RA SF (false discovery rate (FDR) < 0.05). Metabolites of glycolysis
and the tricarboxylic acid cycle were lower in RA compared to OA;
these results concur with higher levels of inflammation, synovial
proliferation and hypoxia found in RA compared to OA. Elevated taurine
in OA may indicate increased subchondral bone sclerosis. We demonstrate
that quantifiable differences in metabolite abundance can be measured
in low volumes of SF by 1H NMR spectroscopy, which may
be clinically useful to aid diagnosis and improve understanding of
disease pathogenesis.
Collapse
Affiliation(s)
- James R Anderson
- Institute of Ageing and Chronic Disease , University of Liverpool , Liverpool L7 8TX , U.K
| | - Susama Chokesuwattanaskul
- Institute of Integrative Biology , University of Liverpool , Liverpool L69 7ZB , U.K.,Chulalongkorn University , Bangkok 10330 , Thailand
| | - Marie M Phelan
- Institute of Integrative Biology , University of Liverpool , Liverpool L69 7ZB , U.K.,HLS Technology Directorate , University of Liverpool , Liverpool L7 8TX , U.K
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery , Maastricht University Medical Centre , 6229 HX Maastricht , The Netherlands
| | - Lu-Yun Lian
- Institute of Integrative Biology , University of Liverpool , Liverpool L69 7ZB , U.K
| | - Mandy J Peffers
- Institute of Ageing and Chronic Disease , University of Liverpool , Liverpool L7 8TX , U.K
| | - Helen L Wright
- Institute of Ageing and Chronic Disease , University of Liverpool , Liverpool L7 8TX , U.K
| |
Collapse
|
34
|
Falconer J, Murphy AN, Young S, Clark AR, Tiziani S, Guma M, Buckley CD. Review: Synovial Cell Metabolism and Chronic Inflammation in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:984-999. [PMID: 29579371 PMCID: PMC6019623 DOI: 10.1002/art.40504] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 03/15/2018] [Indexed: 12/17/2022]
Abstract
Metabolomic studies of body fluids show that immune-mediated inflammatory diseases such as rheumatoid arthritis (RA) are associated with metabolic disruption. This is likely to reflect the increased bioenergetic and biosynthetic demands of sustained inflammation and changes in nutrient and oxygen availability in damaged tissue. The synovial membrane lining layer is the principal site of inflammation in RA. Here, the resident cells are fibroblast-like synoviocytes (FLS) and synovial tissue macrophages, which are transformed toward overproduction of enzymes that degrade cartilage and bone and cytokines that promote immune cell infiltration. Recent studies have shown metabolic changes in both FLS and macrophages from RA patients, and these may be therapeutically targetable. However, because the origins and subset-specific functions of synoviocytes are poorly understood, and the signaling modules that control metabolic deviation in RA synovial cells are yet to be explored, significant additional research is needed to translate these findings to clinical application. Furthermore, in many inflamed tissues, different cell types can forge metabolic collaborations through solute carriers in their membranes to meet a high demand for energy or biomolecules. Such relationships are likely to exist in the synovium and have not been studied. Finally, it is not yet known whether metabolic change is a consequence of disease or whether primary changes to cellular metabolism might underlie or contribute to the pathogenesis of early-stage disease. In this review article, we collate what is known about metabolism in synovial tissue cells and highlight future directions of research in this area.
Collapse
Affiliation(s)
- Jane Falconer
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Anne N Murphy
- Pharmacology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Stephen Young
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Andrew R Clark
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Stefano Tiziani
- Department of Nutritional Sciences & Dell Pediatric Research Institute, University of Texas at Austin, 1400 Barbara Jordan Blvd., Austin, TX
| | - Monica Guma
- Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Christopher D Buckley
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford. UK
| |
Collapse
|
35
|
de Visser HM, Mastbergen SC, Ravipati S, Welsing PMJ, Pinto FC, Lafeber FPJG, Chapman V, Barrett DA, Weinans H. Local and systemic inflammatory lipid profiling in a rat model of osteoarthritis with metabolic dysregulation. PLoS One 2018; 13:e0196308. [PMID: 29684084 PMCID: PMC5912715 DOI: 10.1371/journal.pone.0196308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/10/2018] [Indexed: 01/15/2023] Open
Abstract
Objective Bioactive oxidised lipids (oxylipins) are important signalling mediators, capable of modulating the inflammatory state of the joint and anticipated to be of importance in joint homeostasis and status of osteoarthritis. The aim of this study was to quantify oxylipin levels in plasma and synovial fluid from rats with experimentally induced osteoarthritis to investigate the potential role of oxylipins as a marker in the disease process of early osteoarthritis. Design Forty rats were randomly allocated to a standard or high-fat diet group. After 12 weeks, local cartilage damage was induced in one knee joint in 14 rats of each diet group. The remaining 6 rats per group served as controls. At week 24, samples were collected. Oxylipin levels were quantified by liquid chromatography–mass spectrometry. Results Overall, 31 lipid-derived inflammatory mediators were detected in fasted plasma and synovial fluid. Principal component analysis identified four distinct clusters associated with histopathological changes. Diet induced differences were evident for 13 individual plasma oxylipins, as well as 5,6-EET in synovial fluid. Surgical-model induced differences were evident for three oxylipins in synovial fluid (15-HETE, 8,9-DHET and 17R-ResolvinD1) with a different response in lipid concentrations for synovial fluid and plasma. Conclusions We demonstrate the quantification of oxidised lipids in rat plasma and synovial fluid in a model of early experimental osteoarthritis. Oxylipins in the synovial fluid that were altered as consequence of the surgically induced osteoarthritis were not represented in the plasma. Our findings suggest differential roles of the oxylipins in the local versus peripheral compartment.
Collapse
Affiliation(s)
- H. M. de Visser
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S. C. Mastbergen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| | - S. Ravipati
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
| | - P. M. J. Welsing
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - F. C. Pinto
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
| | - F. P. J. G. Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - V. Chapman
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
- Arthritis Research UK Pain Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kindom
| | - D. A. Barrett
- School of Pharmacy (DAB, FCP) and School of Life Sciences (VC), University of Nottingham, Nottingham, United Kingdom
| | - H. Weinans
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
36
|
Biomarker-guided stratification of autoimmune patients for biologic therapy. Curr Opin Immunol 2017; 49:56-63. [DOI: 10.1016/j.coi.2017.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/22/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
|
37
|
Romão VC, Vital EM, Fonseca JE, Buch MH. Right drug, right patient, right time: aspiration or future promise for biologics in rheumatoid arthritis? Arthritis Res Ther 2017; 19:239. [PMID: 29065909 PMCID: PMC5655983 DOI: 10.1186/s13075-017-1445-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Individualising biologic disease-modifying anti-rheumatic drugs (bDMARDs) to maximise outcomes and deliver safe and cost-effective care is a key goal in the management of rheumatoid arthritis (RA). Investigation to identify predictive tools of bDMARD response is a highly active and prolific area of research. In addition to clinical phenotyping, cellular and molecular characterisation of synovial tissue and blood in patients with RA, using different technologies, can facilitate predictive testing. This narrative review will summarise the literature for the available bDMARD classes and focus on where progress has been made. We will also look ahead and consider the increasing use of 'omics' technologies, the potential they hold as well as the challenges, and what is needed in the future to fully realise our ambition of personalised bDMARD treatment.
Collapse
Affiliation(s)
- Vasco C. Romão
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Department of Rheumatology, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Av. Professor Egas Moniz, 1649-035 Lisboa, Portugal
| | - Edward M. Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - João Eurico Fonseca
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
- Department of Rheumatology, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Av. Professor Egas Moniz, 1649-035 Lisboa, Portugal
| | - Maya H. Buch
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
38
|
Pickens CA, Sordillo LM, Zhang C, Fenton JI. Obesity is positively associated with arachidonic acid-derived 5- and 11-hydroxyeicosatetraenoic acid (HETE). Metabolism 2017; 70:177-191. [PMID: 28403941 DOI: 10.1016/j.metabol.2017.01.034] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Oxylipids are oxygenated polyunsaturated fatty acid (PUFA) metabolites that are responsible for the onset and resolution of the inflammatory response. Enzymatic oxygenation through the lipoxygenase (LOX) or cytochrome P450 (CYP) pathways can form oxylipids that have either proinflammatory or proresolving functions depending on the type of PUFA substrate and degree of metabolism. The objective of this study was to determine how PUFA substrates and their corresponding oxylipids are associated with obesity. METHODS Plasma non-esterified FA and oxylipids were isolated from 123 Caucasian males using solid phase extraction and quantified using high performance liquid chromatography-tandem mass spectrometry. Statistical analyses included linear regressions and polytomous logistic regressions, and the responses were body mass index (BMI) and waist circumference (WC), and serum leptin, total adiponectin, interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and C-peptide. Models were adjusted for age and smoking, and p-values were corrected for false discovery per Benjamini-Hochberg and Bonferroni. RESULTS We report that BMI, WC, and several serum cytokines were highly associated arachidonic acid (ARA)-derived hydroxyeicosatetraenoic acids (HETEs), and vicinal diols (i.e., alcohols on adjacent carbon atoms) derived from several PUFAs. There was a significant linear relationship between BMI, WC, and serum leptin, and ARA-derived 5-, 11-, and 15-HETE. Specifically, BMI and WC were positively associated with proinflammatory 5- and 11-hydroxyeicosatetraenoic acid (HETE), even after normalization to ARA concentrations and false discovery p-value correction. Individuals with 5-HETE concentrations >5.01nmol/L or 11-HETE concentrations and >0.89nmol/L were over 5 times more likely to be obese compared to those with ≤1.86nmol/L and ≤0.39nmol/L, respectively. Vicinal diols from linoleic, eicosapentaenoic, and docosahexaenoic acid were inversely associated with obesity. Across all statistical tests, vicinal diols were inversely associated with obesity whether normalized to parent PUFA concentrations or normalized to precursor epoxides. Interestingly, the proinflammatory cytokines IL-6 and TNF-α were not associated with any oxylipids. Since 5-HETE is a 5LOX product, 11-HETE is marker of lipid peroxidation, and vicinal diols are formed through soluble epoxide hydrolase (sEH) metabolism of CYP epoxygenated PUFAs, therefore, these results indicate that obesity is likely associated with altered metabolism with distinct oxygenating pathways. Taken together, our results indicate that obesity is associated with specific oxylipids indicative of altered PUFA metabolism through several pathways (i.e., LOX, reactive oxygen species, and sEH and CYP epoxygenase), rather than attributed solely to altered dietary PUFA intake.
Collapse
Affiliation(s)
| | | | - Chen Zhang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, East Lansing, MI, United States.
| |
Collapse
|
39
|
Fu J, Cuppen BVJ, Welsing PMJ, van Wietmarschen H, Harms AC, Berger R, Koval S, Fritsch-Stork RDE, Bijlsma JWJ, Hankemeier T, van der Greef J, Lafeber FPJG. Differences between serum polar lipid profiles of male and female rheumatoid arthritis patients in response to glucocorticoid treatment. Inflammopharmacology 2016; 24:397-402. [PMID: 27682325 PMCID: PMC5119840 DOI: 10.1007/s10787-016-0284-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/21/2016] [Indexed: 01/18/2023]
Abstract
Objective As there are pharmacological differences between males and females, and glucocorticoid (GC) treatment is associated with increased cardiovascular mortality rate in rheumatoid arthritis (RA) patients, it is important to study serum polar lipid profiles of male and female patients in response to GC therapy. Gender differences may require an adjustment to the treatment strategy for a selection of patients. Methods Serum samples from 281 RA patients were analysed using a targeted lipidomics platform. The differences in GC use and gender on polar lipid profiles were cross sectionally examined by multiple linear regressions, while correcting for confounding factors. Results Differences in polar lipids between GC users and non-GC users in females and males were merely restricted to lysophospholipids (lysophosphatidylcholines and lysophosphatidylethanolamines). Lysophospholipids in female patients treated with GCs were significantly higher than female patients not treated with GCs (p = 6.0 E−6), whereas no significant difference was observed in male GC users versus non-users (p = 0.397). Conclusion The lysophospholipid profiles in response to GCs were significantly different between male and female RA patients, which may have implications for the cardiovascular risk of GC treatment. Electronic supplementary material The online version of this article (doi:10.1007/s10787-016-0284-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junzeng Fu
- Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands. .,Sino-Dutch Center for Preventive and Personalized Medicine, P.O. Box 360, 3700 AJ, Zeist, The Netherlands.
| | - Bart V J Cuppen
- Rheumatology and Clinical Immunology, University Medical Center Utrecht, F02.127, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Paco M J Welsing
- Rheumatology and Clinical Immunology, University Medical Center Utrecht, F02.127, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Herman van Wietmarschen
- Sino-Dutch Center for Preventive and Personalized Medicine, P.O. Box 360, 3700 AJ, Zeist, The Netherlands
| | - Amy C Harms
- Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Netherlands Metabolomics Center, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Ruud Berger
- Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Netherlands Metabolomics Center, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Slavik Koval
- Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Netherlands Metabolomics Center, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Ruth D E Fritsch-Stork
- Rheumatology and Clinical Immunology, University Medical Center Utrecht, F02.127, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.,1st Medical Department and Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, Hanusch Hospital, Heinrich-Collin-Straße 30, 1140, Vienna, Austria.,Sigmund Freud University, Freudplatz 1, 1020, Vienna, Austria
| | - Johannes W J Bijlsma
- Rheumatology and Clinical Immunology, University Medical Center Utrecht, F02.127, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Thomas Hankemeier
- Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Netherlands Metabolomics Center, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Jan van der Greef
- Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Sino-Dutch Center for Preventive and Personalized Medicine, P.O. Box 360, 3700 AJ, Zeist, The Netherlands.,TNO, Netherlands Organization for Applied Scientific Research, Microbiology and Systems Biology, Zeist, The Netherlands.,Netherlands Metabolomics Center, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Floris P J G Lafeber
- Rheumatology and Clinical Immunology, University Medical Center Utrecht, F02.127, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|