1
|
Caso F, Costa L, Megna M, Cascone M, Maione F, Giacomelli R, Scarpa R, Ruscitti P. Early psoriatic arthritis: clinical and therapeutic challenges. Expert Opin Investig Drugs 2024; 33:945-965. [PMID: 39041193 DOI: 10.1080/13543784.2024.2383421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic immunoinflammatory disease of the enthesis and adjacent synovium, skin, and nail, which early diagnosis may be crucial for starting a prompt therapeutic intervention. Theoretically, early treatment offers the advantage of acting on the reduction of the articular damage progression since initial phases of the disease. AREAS COVERED This review explores the challenges of clinical-diagnostic aspects and the underlying pathophysiology of early PsA phases, as well as the evidence evaluating the impact of early intervention on disease outcomes. EXPERT OPINION Main instruments for early PsA diagnosis include recognizing synovial-entheseal inflammatory signs at onset, improving screening PsA high-risk subjects, and increasing disease knowledge of physicians and patients with psoriasis or familial history. PsA continues to significantly impact on the Quality of Life of patients affected by the disease, making necessary to deeply study clinical manifestations, risk factors, and underlying immunoinflammatory mechanisms, as well as to identify biomarkers for early identification. Additionally, it remains a need to increase more evidence on understanding how early treatment of PsA and of psoriasis might influence the course of the disease.
Collapse
Affiliation(s)
- Francesco Caso
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Matteo Megna
- Section of Dermatology - Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli, Italy
| | - Mario Cascone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Roberto Giacomelli
- Research and Clinical Unit of immunorheumatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Rheumatology, Immunology and Clinical Medicine Unit, Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
2
|
Pinto-Tasende JA, Fernandez-Moreno M, Rego Perez I, Fernandez-Lopez JC, Oreiro-Villar N, De Toro Santos FJ, Blanco-García FJ. Higher Synovial Immunohistochemistry Reactivity of IL-17A, Dkk1, and TGF-β1 in Patients with Early Psoriatic Arthritis and Rheumatoid Arthritis Could Predict the Use of Biologics. Biomedicines 2024; 12:815. [PMID: 38672170 PMCID: PMC11048598 DOI: 10.3390/biomedicines12040815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Delay in diagnosis and therapy in patients with arthritis commonly leads to progressive articular damage. The study aimed to investigate the immunohistochemical reactivity of synovial cytokines associated with inflammation and the bone erosives/neoformatives processes among individuals diagnosed with psoriatic arthritis (PsA), rheumatoid arthritis (RA), osteoarthritis (OA), and radiographic axial spondyloarthritis (r-axSpA), with the intention of identifying potential biomarkers. METHODS Specimens were collected from the inflamed knee joints of patients referred for arthroscopic procedures, and the synovial tissue (ST) was prepared for quantifying protein expression through immunohistochemical analysis (% expressed in Ratio_Area-Intensity) for TGF-β1, IL-17A, Dkk1, BMP2, BMP4, and Wnt5b. The collected data underwent thorough analysis and examination of their predictive capabilities utilising receiver operating characteristic (ROC) curves. RESULTS Valid synovial tissue samples were acquired from 40 patients for IHC quantification analysis. Initially, these patients had not undergone treatment with biologics. However, after 5 years, 4 out of 13 patients diagnosed with PsA and two out of nine patients diagnosed with RA had commenced biologic treatments. Individuals with early PsA who received subsequent biologic treatment exhibited significantly elevated IHC reactivity in ST for TGF-β1 (p = 0.015). Additionally, patients with both PsA and RA who underwent biologic therapy displayed increased IHC reactivity for IL-17A (p = 0.016), TGF-β1 (p = 0.009), and Dkk1 (p = 0.042). ROC curve analysis of IHC reactivity for TGF-β1, Dkk1, and IL-17A in the synovial seems to predict future treatment with biologics in the next 5 years with the area under the curve (AUC) of a combined sum of the three values: AUC: 0.828 (95% CI: 0.689-0.968; p 0.005) S 75% E 84.4%. CONCLUSIONS Higher synovial immunohistochemistry reactivity of IL-17A, Dkk1, and TGF-β1 in patients with early psoriatic arthritis and rheumatoid arthritis may serve as potential indicators for predicting the necessity of utilising biologic treatments.
Collapse
Affiliation(s)
- Jose A. Pinto-Tasende
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - Mercedes Fernandez-Moreno
- Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain; (M.F.-M.); (I.R.P.)
| | - Ignacio Rego Perez
- Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, 15006 A Coruña, Spain; (M.F.-M.); (I.R.P.)
| | - J. Carlos Fernandez-Lopez
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - Natividad Oreiro-Villar
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - F. Javier De Toro Santos
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| | - Francisco J. Blanco-García
- Department of Rheumatology, Institute of Biomedical Research of A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña, Universidade de A Coruña, 15006 A Coruña, Spain; (J.C.F.-L.); (N.O.-V.); (F.J.D.T.S.); (F.J.B.-G.)
| |
Collapse
|
3
|
Skougaard M, Søndergaard MF, Ditlev SB, Kristensen LE. Changes in Inflammatory Cytokines in Responders and Non-Responders to TNFα Inhibitor and IL-17A Inhibitor: A Study Examining Psoriatic Arthritis Patients. Int J Mol Sci 2024; 25:3002. [PMID: 38474247 PMCID: PMC10932211 DOI: 10.3390/ijms25053002] [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: 01/30/2024] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
This study aimed to examine the changes in biomarker levels in responders and non-responders to tumor necrosis factor alpha inhibitor (TNFi) and interleukin-17A inhibitor (IL-17Ai) in psoriatic arthritis (PsA) patients over a 4-month period after treatment initiation. A total of 68 PsA patients initiating either TNFi, IL-17Ai, or methotrexate treatment were included. Blood plasma and clinical outcome measures were collected adjacent to treatment initiation and after four months. A commercially available multiplex immunoassay was included to evaluate 54 biomarkers. Mean changes were used to evaluate change over time. A statistically significant decrease in pro-inflammatory cytokines IL-6 (log-transformed mean change -0.97, 95%CI -4.30; 2.37, [p = 0.032]) and an increase in anti-inflammatory IL-10 (0.38, 95%CI 1.74; 2.50 [p = 0.010]) were seen in TNFi responders. Meanwhile, a statistically significant increase in the target cytokine IL-17A was seen in both IL-17Ai responders (2.49, 95%CI -1.84; 6.85 [p = 0.031]) and non-responders (2.48, 95%CI -1.46; 6.41 [p = 0.001]). This study demonstrated differing changes in cytokine levels when comparing treatment responders and non-responders, highlighting the need to improve the understanding of the different immune response mechanisms explaining different responses to medical treatment in PsA patients.
Collapse
Affiliation(s)
- Marie Skougaard
- The Parker Institute, Copenhagen University Hospital Bispebjerg and Frederiksberg, Nordre Fasanvej 57, 2000 Frederiksberg, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Magnus Friis Søndergaard
- Copenhagen Center for Translational Research, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational Research, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Lars Erik Kristensen
- The Parker Institute, Copenhagen University Hospital Bispebjerg and Frederiksberg, Nordre Fasanvej 57, 2000 Frederiksberg, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
4
|
Tanaka Y, Taylor PC, Elboudwarej E, Hertz A, Shao X, Malkov VA, Matsushima H, Emoto K, Downie B, Takeuchi T. Filgotinib Modulates Inflammation-Associated Peripheral Blood Protein Biomarkers in Adults with Active Rheumatoid Arthritis and Prior Inadequate Response to Methotrexate. Rheumatol Ther 2023; 10:1335-1348. [PMID: 37490202 PMCID: PMC10468462 DOI: 10.1007/s40744-023-00583-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/28/2023] [Indexed: 07/26/2023] Open
Abstract
INTRODUCTION Our aim was to evaluate protein biomarker changes related to the administration of filgotinib, a Janus kinase (JAK) 1 preferential inhibitor, in patients with moderately to severely active rheumatoid arthritis (RA) with inadequate response to methotrexate. METHODS Plasma and serum samples were collected from patients enrolled in FINCH 1 (NCT02889796), a Phase 3 trial. Patients with stable backgrounds of methotrexate were randomly assigned once-daily oral filgotinib 200 or 100 mg, subcutaneous adalimumab 40 mg every 2 weeks (W), or placebo. Up to 35 biomarkers were analyzed at baseline, W4, and W12 with enzyme-linked immunosorbent assays and chemiluminescence and electrochemiluminescence assays. RESULTS At baseline, four distinct biomarker clusters were identified. The strongest intragroup correlations were in bone-cartilage resorption/inflammation and JAK/signal transducer and activator of transcription (STAT) signaling activity. At baseline, significant positive correlations were identified for cytokines with patient-reported pain and with patient measures of fatigue. Filgotinib reduced levels of cytokines associated with inflammation and cell migration as early as W4 and through W12. Compared to adalimumab, filgotinib induced significant reductions in bone-related turnover biomarkers, N-telopeptide of type 1 collagen and C-telopeptide 1, as well as biomarkers associated with baseline disease activity. No baseline predictors of therapeutic response to filgotinib were identified. CONCLUSIONS Filgotinib reduced peripheral protein biomarkers associated with JAK/STAT signaling, inflammatory signaling, immune cell migration, and bone resorption as soon as W4 in FINCH 1. Effects were dose-dependent and consistent with the clinical efficacy of filgotinib observed in FINCH 1. The changes in peripheral biomarkers associated with filgotinib treatment in methotrexate-experienced patients are consistent with changes observed in both methotrexate-naïve and biologic disease-modifying antirheumatic drug-experienced RA populations. These data demonstrate dose-dependent effects of preferential JAK1 inhibition by filgotinib on peripheral blood protein biomarkers in methotrexate-experienced patients with RA. TRIAL REGISTRATION ClinicalTrials.gov, NCT02889796.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Kitakyushu, 807-8555, Japan.
| | - Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Windmill Road, Headington, Oxford, OX3 7LD, UK.
| | | | | | | | | | | | | | | | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
- Saitama Medical University, Saitama, Japan
| |
Collapse
|
5
|
Riitano G, Recalchi S, Capozzi A, Manganelli V, Misasi R, Garofalo T, Sorice M, Longo A. The Role of Autophagy as a Trigger of Post-Translational Modifications of Proteins and Extracellular Vesicles in the Pathogenesis of Rheumatoid Arthritis. Int J Mol Sci 2023; 24:12764. [PMID: 37628944 PMCID: PMC10454292 DOI: 10.3390/ijms241612764] [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: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease, characterized by persistent joint inflammation, leading to cartilage and bone destruction. Autoantibody production is directed to post-translational modified (PTM) proteins, i.e., citrullinated or carbamylated. Autophagy may be the common feature in several types of stress (smoking, joint injury, and infections) and may be involved in post-translational modifications (PTMs) in proteins and the generation of citrullinated and carbamylated peptides recognized by the immune system in RA patients, with a consequent breakage of tolerance. Interestingly, autophagy actively provides information to neighboring cells via a process called secretory autophagy. Secretory autophagy combines the autophagy machinery with the secretion of cellular content via extracellular vesicles (EVs). A role for exosomes in RA pathogenesis has been recently demonstrated. Exosomes are involved in intercellular communications, and upregulated proteins and RNAs may contribute to the development of inflammatory arthritis and the progression of RA. In RA, most of the exosomes are produced by leukocytes and synoviocytes, which are loaded with PTM proteins, mainly citrullinated proteins, inflammatory molecules, and enzymes that are implicated in RA pathogenesis. Microvesicles derived from cell plasma membrane may also be loaded with PTM proteins, playing a role in the immunopathogenesis of RA. An analysis of changes in EV profiles, including PTM proteins, could be a useful tool for the prevention of inflammation in RA patients and help in the discovery of personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maurizio Sorice
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (S.R.); (A.C.); (V.M.); (R.M.); (T.G.); (A.L.)
| | | |
Collapse
|
6
|
Zhou W, Jia J, Qu HQ, Ma F, Li J, Qi X, Meng X, Ding Z, Zheng G, Hakonarson H, Zeng X, Li J, Xia Q. Identification of copy number variants contributing to hallux valgus. Front Genet 2023; 14:1116284. [PMID: 37035746 PMCID: PMC10076598 DOI: 10.3389/fgene.2023.1116284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/13/2023] [Indexed: 04/11/2023] Open
Abstract
Hallux valgus is a common form of foot deformity, and genetic factors contribute substantially to the pathogenesis of hallux valgus deformity. We conducted a genetic study on the structural variants underlying familial hallux valgus using whole exome sequencing approach. Twenty individuals from five hallux valgus families and two sporadic cases were included in this study. A total of 372 copy number variations were found and passed quality control filtering. Among them, 43 were only present in cases but not in controls or healthy individuals in the database of genomic variants. The genes covered by these copy number variations were enriched in gene sets related to immune signaling pathway, and cytochrome P450 metabolism. The hereditary CNVs demonstrate a dominant inheritance pattern. Two candidate pathogenic CNVs were further validated by quantitative-PCR. This study suggests that hallux valgus is a degenerative joint disease involving the dysregulation of immune and metabolism signaling pathways.
Collapse
Affiliation(s)
- Wentao Zhou
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jun Jia
- Department of Surgery of Foot and Ankle, Tianjin Hospital, Tianjin, China
| | - Hui-Qi Qu
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Feier Ma
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junyi Li
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohui Qi
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xinyi Meng
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, China
| | - Gang Zheng
- National Supercomputer Center in Tianjin (NSCC-TJ), Tianjin, China
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiantie Zeng
- Department of Surgery of Foot and Ankle, Tianjin Hospital, Tianjin, China
| | - Jin Li
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qianghua Xia
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- *Correspondence: Qianghua Xia,
| |
Collapse
|
7
|
Zheng K, Bai J, Yang H, Xu Y, Pan G, Wang H, Geng D. Nanomaterial-assisted theranosis of bone diseases. Bioact Mater 2022; 24:263-312. [PMID: 36632509 PMCID: PMC9813540 DOI: 10.1016/j.bioactmat.2022.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/27/2022] Open
Abstract
Bone-related diseases refer to a group of skeletal disorders that are characterized by bone and cartilage destruction. Conventional approaches can regulate bone homeostasis to a certain extent. However, these therapies are still associated with some undesirable problems. Fortunately, recent advances in nanomaterials have provided unprecedented opportunities for diagnosis and therapy of bone-related diseases. This review provides a comprehensive and up-to-date overview of current advanced theranostic nanomaterials in bone-related diseases. First, the potential utility of nanomaterials for biological imaging and biomarker detection is illustrated. Second, nanomaterials serve as therapeutic delivery platforms with special functions for bone homeostasis regulation and cellular modulation are highlighted. Finally, perspectives in this field are offered, including current key bottlenecks and future directions, which may be helpful for exploiting nanomaterials with novel properties and unique functions. This review will provide scientific guidance to enhance the development of advanced nanomaterials for the diagnosis and therapy of bone-related diseases.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author.Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China,Corresponding author.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author. Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
8
|
Kovács OT, Tóth E, Ozohanics O, Soltész-Katona E, Marton N, Buzás EI, Hunyady L, Drahos L, Turu G, Nagy G. Proteomic Changes of Osteoclast Differentiation in Rheumatoid and Psoriatic Arthritis Reveal Functional Differences. Front Immunol 2022; 13:892970. [PMID: 35860269 PMCID: PMC9289121 DOI: 10.3389/fimmu.2022.892970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundOsteoclasts play a crucial role in the maintenance, repair, and remodeling of bones of the adult vertebral skeleton due to their bone resorption capability. Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are associated with increased activity of osteoclasts.ObjectivesOur study aimed to investigate the dynamic proteomic changes during osteoclast differentiation in healthy donors, in RA, and PsA.MethodsBlood samples of healthy donors, RA, and PsA patients were collected, and monocytes were isolated and differentiated into osteoclasts in vitro using macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor κB ligand (RANK-L). Mass spectrometry-based proteomics was used to analyze proteins from cell lysates. The expression changes were analyzed with Gene Set Enrichment Analysis (GSEA).ResultsThe analysis of the proteomic changes revealed that during the differentiation of the human osteoclasts, expression of the proteins involved in metabolic activity, secretory function, and cell polarity is increased; by contrast, signaling pathways involved in the immune functions are downregulated. Interestingly, the differences between cells of healthy donors and RA/PsA patients are most pronounced after the final steps of differentiation to osteoclasts. In addition, both in RA and PsA the differentiation is characterized by decreased metabolic activity, associated with various immune pathway activities; furthermore by accelerated cytokine production in RA.ConclusionsOur results shed light on the characteristic proteomic changes during human osteoclast differentiation and expression differences in RA and PsA, which reveal important pathophysiological insights in both diseases.
Collapse
Affiliation(s)
- Orsolya Tünde Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Eszter Tóth
- Institute of Organic Chemistry, Eötvös Loránd Research Network, Research Centre for Natural Sciences, Budapest, Hungary
| | - Olivér Ozohanics
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Eszter Soltész-Katona
- Department of Physiology, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Laboratory of Molecular Physiology, Eötvös Loránd Research Network, Budapest, Hungary
| | - Nikolett Marton
- Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Edit Irén Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Immune-Proteogenomics Research Group, Budapest, Hungary
- Hungarian Centre of Excellence for Molecular Medicine - Semmelweis University (HCEMM-SU) Extracellular Vesicles Research Group, Budapest, Hungary
| | - László Hunyady
- Department of Physiology, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Laboratory of Molecular Physiology, Eötvös Loránd Research Network, Budapest, Hungary
- Institute of Enzymology, Eötvös Loránd Research Network, Research Centre for Natural Sciences, Budapest, Hungary
| | - László Drahos
- Institute of Organic Chemistry, Eötvös Loránd Research Network, Research Centre for Natural Sciences, Budapest, Hungary
| | - Gábor Turu
- Department of Physiology, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Laboratory of Molecular Physiology, Eötvös Loránd Research Network, Budapest, Hungary
- *Correspondence: Gábor Turu,
| | - György Nagy
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| |
Collapse
|
9
|
Durez P, Westhovens R, Baeke F, Elbez Y, Robert S, Ahmad HA. Identification of poor prognostic joint locations in an early rheumatoid arthritis cohort at risk of rapidly progressing disease: a post-hoc analysis of the Phase III AGREE study. BMC Rheumatol 2022; 6:24. [PMID: 35418172 PMCID: PMC9009012 DOI: 10.1186/s41927-022-00252-4] [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: 08/20/2021] [Accepted: 02/09/2022] [Indexed: 11/23/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a heterogeneous disease with established poor prognostic factors such as seropositivity, joint damage, and high disease activity at an early, treatment-naïve stage of disease. However, few studies have examined if specific joint locations are correlated with these factors in such a population. This analysis explored the potential correlation of individual swollen and erosive joints with other disease characteristics at baseline and with remission rates in a post-hoc analysis of the Phase III randomized AGREE study. Methods Methotrexate (MTX)-naïve, erosive, RF- and/or ACPA-positive early RA patients (N = 509) were retrospectively evaluated. Baseline joint swelling was analyzed for large and small joints. Baseline erosions were analyzed for wrist, MCP1–5, IP1, PIP2–5 and MTP1–5. Remission rates were assessed after 6 months of treatment with abatacept (ABA) + MTX (N = 256) or MTX (N = 253). The following statistical tests were used: Chi-Square or Fisher’s exact test (categorical variables); Student’s t-test or Wilcoxon rank-sum test (continuous variables); continuity-corrected Chi-square test (efficacy remission endpoints). Results Baseline swelling was most frequent in wrist (91.9%) and MCP2 joint (89.1%), while baseline erosion was most frequent in MTP5 joint (43.5%). Swollen shoulder was significantly correlated (p < 0.0001) with swelling of almost all other large or medium joints. Baseline swelling in the knee, temporomandibular joint (TMJ), wrist and elbow was highly correlated (p < 0.001) with higher tender and swollen joint counts, higher DAS28(CRP) and higher SDAI and CDAI. Baseline swelling was not correlated with erosion per joint, except for MCP2. The largest difference in mean Boolean remission rates at 6 months was in patients with baseline swollen wrist favoring ABA + MTX (14.0% vs 4.4%; p < 0.001). Conclusions Swelling in the large and medium joints (knee, TMJ, elbow and wrist) was highly correlated with severe disease activity while MCP2 swelling seemed to be correlated with joint damage. The correlation of joint locations at an early, treatment-naïve stage with poor prognostic factors, higher disease activity and joint damage, could establish a rapidly progressing anatomical pattern in early RA. Trial registration: ClinicalTrials.gov NCT00122382, registered July 2005. Supplementary Information The online version contains supplementary material available at 10.1186/s41927-022-00252-4.
Collapse
Affiliation(s)
- Patrick Durez
- Institut de Recherche Expérimentale Et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Service de rhumatologie, 1200, Bruxelles, Belgium.
| | - Rene Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Femke Baeke
- Bristol-Myers Squibb, Braine L'Alleud, Belgium
| | | | | | | |
Collapse
|
10
|
Lättekivi F, Guljavina I, Midekessa G, Viil J, Heath PR, Bæk R, Jørgensen MM, Andronowska A, Kingo K, Fazeli A. Profiling Blood Serum Extracellular Vesicles in Plaque Psoriasis and Psoriatic Arthritis Patients Reveals Potential Disease Biomarkers. Int J Mol Sci 2022; 23:ijms23074005. [PMID: 35409365 PMCID: PMC9000144 DOI: 10.3390/ijms23074005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/15/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Psoriasis vulgaris (PsV) and psoriatic arthritis (PsA) are inflammatory diseases with unresolved pathophysiological aspects. Extracellular vesicles (EVs) play an important role in intercellular communication. We compared the miRNA contents and surface proteome of the EVs in the blood serum of PsV and PsA patients to healthy controls. Size-exclusion chromatography was used to isolate EVs from the blood serum of 12 PsV patients, 12 PsA patients and 12 healthy control subjects. EV samples were characterized and RNA sequencing was used to identify differentially enriched EV-bound miRNAs. We found 212 differentially enriched EV-bound miRNAs present in both PsV and PsA groups—a total of 13 miRNAs at FDR ≤ 0.05. The predicted target genes of these miRNAs were significantly related to lesser known but potentially disease-relevant pathways. The EV array revealed that PsV patient EV samples were significantly enriched with CD9 EV-marker compared to controls. Analysis of EV-bound miRNAs suggests that signaling via EVs in the blood serum could play a role in the pathophysiological processes of PsV and PsA. EVs may be able to fill the void in clinically applicable diagnostic and prognostic biomarkers for PsV and PsA.
Collapse
Affiliation(s)
- Freddy Lättekivi
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia; (F.L.); (I.G.); (G.M.)
| | - Irina Guljavina
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia; (F.L.); (I.G.); (G.M.)
| | - Getnet Midekessa
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia; (F.L.); (I.G.); (G.M.)
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia
| | - Janeli Viil
- Department of Pharmacology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia;
| | - Paul R. Heath
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK;
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, Urbansgade 32-36, 9000 Aalborg, Denmark; (R.B.); (M.M.J.)
| | - Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Urbansgade 32-36, 9000 Aalborg, Denmark; (R.B.); (M.M.J.)
- Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, 9220 Aalborg, Denmark
| | - Aneta Andronowska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima St. 10, 10-748 Olsztyn, Poland;
| | - Kulli Kingo
- Clinic of Dermatology, Institute of Clinical Medicine, University of Tartu, Raja 31, 50417 Tartu, Estonia;
- Clinic of Dermatology, Tartu University Hospital, Raja 31, 50417 Tartu, Estonia
| | - Alireza Fazeli
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 14b, 50411 Tartu, Estonia; (F.L.); (I.G.); (G.M.)
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 62, 51006 Tartu, Estonia
- Academic Unit of Reproductive and Developmental Medicine, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield S10 2SF, UK
- Correspondence: ; Tel.: +372-737-4425
| |
Collapse
|
11
|
Marzaioli V, Canavan M, Floudas A, Flynn K, Mullan R, Veale DJ, Fearon U. CD209/CD14 + Dendritic Cells Characterization in Rheumatoid and Psoriatic Arthritis Patients: Activation, Synovial Infiltration, and Therapeutic Targeting. Front Immunol 2022; 12:722349. [PMID: 35095831 PMCID: PMC8789658 DOI: 10.3389/fimmu.2021.722349] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/17/2021] [Indexed: 12/29/2022] Open
Abstract
Dendritic cells (DC) have a key role in the initiation and progression of inflammatory arthritis (IA). In this study, we identified a DC population that derive from monocytes, characterized as CD209/CD14+ DC, expressing classical DC markers (HLADR, CD11c) and the Mo-DC marker (CD209), while also retaining the monocytic marker CD14. This CD209/CD14+ DC population is present in the circulation of Healthy Control (HC), with increased frequency in Rheumatoid Arthritis (RA) and Psoriatic arthritic (PsA) patients. We demonstrate, for the first time, that circulatory IA CD209/CD14+ DC express more cytokines (IL1β/IL6/IL12/TNFα) and display a unique chemokine receptor expression and co-expression profiles compared to HC. We demonstrated that CD209/CD14+ DC are enriched in the inflamed joint where they display a unique inflammatory and maturation phenotype, with increased CD40 and CD80 and co-expression of specific chemokine receptors, displaying unique patterns between PsA and RA. We developed a new protocol of magnetic isolation and expansion for CD209+ DC from blood and identified transcriptional differences involved in endocytosis/antigen presentation between RA and PsA CD209+ DC. In addition, we observed that culture of healthy CD209+ DC with IA synovial fluid (SF), but not Osteoarthritis (OA) SF, was sufficient to induce the development of CD209/CD14+ DC, leading to a poly-mature DC phenotype. In addition, differential effects were observed in terms of chemokine receptor and chemokine expression, with healthy CD209+ DC displaying increased expression/co-expression of CCR6, CCR7, CXCR3, CXCR4 and CXCR5 when cultured with RA SF, while an increase in the chemokines CCR3, CXCL10 and CXCL11 was observed when cultured with PsA SF. This effect may be mediated in part by the observed differential increase in chemokines expressed in RA vs PsA SF. Finally, we observed that the JAK/STAT pathway, but not the NF-κB pathway (driven by TNFα), regulated CD209/CD14+ DC function in terms of activation, inflammatory state, and migratory capacity. In conclusion, we identified a novel CD209/CD14+ DC population, which is active in the circulation of RA and PsA, an effect potentiated once they enter the joint. Furthermore, we demonstrated that JAK/STAT inhibition can be used as a therapeutic strategy to decrease the inflammatory state of the pathogenic CD209/CD14+ DC.
Collapse
Affiliation(s)
- Viviana Marzaioli
- Rheumatology EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St Vincent's University Hospital, University College Dublin, Dublin, Ireland.,Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Keelin Flynn
- Rheumatology EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St Vincent's University Hospital, University College Dublin, Dublin, Ireland.,Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ronan Mullan
- Department of Rheumatology, Tallaght University Hospital, Dublin, Ireland
| | - Douglas J Veale
- Rheumatology EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Rheumatology EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St Vincent's University Hospital, University College Dublin, Dublin, Ireland.,Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Funk PJ, Perche PO, Singh R, Kelly KA, Feldman SR. Comparing available JAK inhibitors for treating patients with psoriasis. Expert Rev Clin Immunol 2022; 18:281-294. [DOI: 10.1080/1744666x.2022.2039121] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Parker J. Funk
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Patrick O. Perche
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Rohan Singh
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Katherine A. Kelly
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Steven R. Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Social Sciences & Health Policy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
13
|
Antirheumatic treatment is associated with reduced serum Syndecan-1 in Rheumatoid Arthritis. PLoS One 2021; 16:e0253247. [PMID: 34242246 PMCID: PMC8270157 DOI: 10.1371/journal.pone.0253247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/01/2021] [Indexed: 12/03/2022] Open
Abstract
The endothelial glycocalyx (EG) is essential for proper function of the endothelium and for vascular integrity, but its role in premature atherogenesis in rheumatoid arthritis (RA) has not been studied yet. EG impairment can play a role in pathogenesis of vascular disease, and one of its characteristics is shedding of syndecan-1 from endothelial cells. Syndecan-1 shedding is mediated by matrix metalloproteinase-9 (MMP-9) and counteracted by tissue inhibitor of metalloproteinases (TIMP)-1. Cardiovascular disease risk in RA is reversible by disease modifying antirheumatic drugs (DMARDs), but the exact modes of action are still unclear. Therefore, we examined effects of DMARDs on syndecan-1, MMP-9 and TIMP-1 in RA patients, and searched for associations between these parameters and inflammatory activity. From the observational PSARA study, we examined 39 patients starting with methotrexate (MTX) monotherapy (in MTX naïve patients, n = 19) or tumor necrosis factor inhibitors (TNFi) in combination with MTX (in MTX non-responders, n = 20) due to active RA. Serum syndecan-1, MMP-9 and TIMP-1 were measured at baseline and after six weeks of treatment. Serum syndecan-1 (p = 0.008) and TIMP-1 (p<0.001) levels decreased after six weeks of anti-rheumatic treatment. Levels of MMP-9 also decreased, but the difference was not statistically significant. The improvement in syndecan-1 levels were independent of changes in inflammatory activity. There was no significant difference in changes in syndecan-1 levels from baseline to 6 weeks between the MTX and TNFi groups, however the change was significant within the MTX group. Six weeks of antirheumatic treatment was associated with reduction in serum levels of syndecan-1, which might reflect reduced syndecan-1 shedding from EG. Thus, it is possible that EG-preserving properties of DMARDs might contribute to their cardioprotective effects. These effects may be at least partly independent of their anti-inflammatory actions. Our findings do not support the notion that syndecan-1 shedding in RA is mediated mainly by increased MMP-9 or decreased TIMP-9 serum concentration.
Collapse
|
14
|
Mc Ardle A, Kwasnik A, Szenpetery A, Hernandez B, Parnell A, de Jager W, de Roock S, FitzGerald O, Pennington SR. Identification and Evaluation of Serum Protein Biomarkers Which Differentiate Psoriatic from Rheumatoid Arthritis. Arthritis Rheumatol 2021; 74:81-91. [PMID: 34114357 DOI: 10.1002/art.41899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/08/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVES To identify serum protein biomarkers which might separate early inflammatory arthritis (EIA) patients with psoriatic arthritis (PsA) from those with rheumatoid arthritis (RA) and may be used to support appropriate early intervention. METHODS The serum proteome of patients with PsA and RA was interrogated using nano-flow liquid chromatography mass spectrometry (nLC-MS/MS) (n=64 patients), an aptamer-based assay (SOMAscan) targeting 1,129 proteins (n=36 patients) and a multiplexed antibody assay (Luminex) for 48 proteins (n=64 patients). Multiple reaction monitoring assays (MRM) were developed to evaluate the performance of putative markers using the discovery cohort (n=60) and subsequently an independent cohort of PsA and RA patients (n=167). RESULTS Multivariate machine learning analysis of the protein discovery data from the three platforms revealed that it was possible to discriminate PsA from RA patients with an area under the curve (AUC) of 0.94 for nLC-MS/MS, 0.69 for bead based immunoassay measurements and 0.73 for aptamer based analysis. Subsequently in the separate verification and evaluation studies, random forest models revealed that a subset of proteins measured by MRM could differentiate PsA and RA patients with AUCs of 0.79 and 0.85 respectively. CONCLUSION We report a serum protein biomarker panel which can separate EIA patients with PsA from those with RA. With continued evaluation and refinement using additional and larger patient cohorts including those with other arthropathies we suggest the panel identified here could contribute toward improved clinical decision making.
Collapse
Affiliation(s)
- Angela Mc Ardle
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Anna Kwasnik
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Agnes Szenpetery
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Belinda Hernandez
- School of Medical Gerontology, TILDA (The Irish Longitudinal Study on Aging), Trinity College Dublin, Ireland.,School of Mathematics and Statistics, University College Dublin, Ireland
| | - Andrew Parnell
- School of Mathematics and Statistics, University College Dublin, Ireland
| | - Wilco de Jager
- Department of Paediatric Immunology, Laboratory of Translation Immunology LTI, Wilhelmina Children Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands.,Multiplex Core Facility, Laboratory of Translational Immunology LTI, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sytze de Roock
- Multiplex Core Facility, Laboratory of Translational Immunology LTI, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Oliver FitzGerald
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Stephen R Pennington
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| |
Collapse
|
15
|
Puentes-Osorio Y, Amariles P, Calleja MÁ, Merino V, Díaz-Coronado JC, Taborda D. Potential clinical biomarkers in rheumatoid arthritis with an omic approach. AUTOIMMUNITY HIGHLIGHTS 2021; 12:9. [PMID: 34059137 PMCID: PMC8165788 DOI: 10.1186/s13317-021-00152-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022]
Abstract
Objective To aid in the selection of the most suitable therapeutic option in patients with diagnosis of rheumatoid arthritis according to the phase of disease, through the review of articles that identify omics biological markers. Methods A systematic review in PubMed/Medline databases was performed. We searched articles from August 2014 to September 2019, in English and Spanish, filtered by title and full text; and using the terms "Biomarkers" AND “Rheumatoid arthritis". Results This article supplies an exhaustive review from research of objective measurement, omics biomarkers and how disease activity appraise decrease unpredictability in treatment determinations, and finally, economic, and clinical outcomes of treatment options by biomarkers’ potential influence. A total of 122 articles were included. Only 92 met the established criteria for review purposes and 17 relevant references about the topic were included as well. Therefore, it was possible to identify 196 potential clinical biomarkers: 22 non-omics, 20 epigenomics, 33 genomics, 21 transcriptomics, 78 proteomics, 4 glycomics, 1 lipidomics and 17 metabolomics. Conclusion A biomarker is a measurable indicator of some, biochemical, physiological, or morphological condition; evaluable at a molecular, biochemical, or cellular level. Biomarkers work as indicators of physiological or pathological processes, or as a result of a therapeutic management. In the last five years, new biomarkers have been identified, especially the omics, which are those that proceed from the investigation of genes (genomics), metabolites (metabolomics), and proteins (proteomics). These biomarkers contribute to the physician choosing the best therapeutic option in patients with rheumatoid arthritis.
Collapse
|
16
|
Malhotra H, Garg V, Singh G. Biomarker Approach Towards Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2021; 17:162-175. [PMID: 33327920 DOI: 10.2174/1573397116666201216164013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis is an auto-immune disorder, recognized by cartilage as well as bone destruction, which causes irreversible joint deformities, which further results in functional limitations in the patient. Genes like HLA-DRB1 and PTPN22 are likely implicated in the genetic predisposition of rheumatoid arthritis pathology. The first and foremost clinical manifestation in a person with rheumatoid arthritis is joint destruction followed by cartilage and bone destruction caused by cell-cell interactions. The cell-cell interactions are thought to be initialized through the contact of antigen-presenting cells (APC) with CD4+ cells, leading to the progression of the disease. APC includes a complex of class ІІ major histocompatibility complex molecules along with peptide antigens and binds to the receptors present on the surface of T-cells. Further, the activation of macrophages is followed by the release of various pro-inflammatory cytokines such as IL-1 and TNF-α, which lead to the secretion of enzymes that degrade proteoglycan and collagen, which in turn, increase tissue degradation. Biomarkers like IL-6, IL-12, IL-8 and IL-18, 14-3-3η, RANKL, IFN-γ, IFN-β and TGF-β have been designated as key biomarkers in disease development and progression. The study of these biomarkers is very important as they act as a molecular indicator of pathological processes that aggravate the disease.
Collapse
Affiliation(s)
- Hitesh Malhotra
- Chandigarh College of Pharmacy Landran, Mohali, Punjab, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
17
|
Belman S, Walsh JA, Carroll C, Milliken M, Haaland B, Duffin KC, Krueger GG, Feng BJ. Psoriasis Characteristics for the Early Detection of Psoriatic Arthritis. J Rheumatol 2021; 48:1559-1565. [PMID: 33858978 DOI: 10.3899/jrheum.201123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Delays in the diagnosis and treatment of psoriatic arthritis (PsA) are common. These delays contribute to impairments in quality of life and joint damage. This study aims to calculate the incidence rate of PsA over time and identify clinical features that may be used for PsA prediction in patients with psoriasis (PsO). METHODS The study population for PsA incidence analysis included 1128 participants enrolled in the Utah Psoriasis Initiative between 2002 and 2014. Clinical evaluation and medical record review were performed to identify new cases of PsA after enrollment. To identify PsO features associated with PsA, the population was restricted to 627 participants who did not have PsA before PsO phenotyping and had been followed up for subsequent PsA diagnosis. We conducted Cox proportional hazard regressions to estimate the HR of PsA associated with PsO characteristics and other health-related features. RESULTS PsA incidence rate increased for > 60 years following PsO onset (trend P < 0.0001). There was a significant association between PsA and induration severity in untreated lesions (P < 0.001, HR 1.46), history of fingernail involvement (P < 0.001, HR 2.38), pustular PsO (P < 0.001, HR 3.32), fingernail involvement at enrollment (P < 0.001, HR 2.04), and Koebner phenomenon (P < 0.001, HR 1.90). Multivariate analysis yielded a model that included a history of fingernail involvement (P < 0.001, HR 2.16) and untreated induration (P < 0.001, HR 1.41). CONCLUSION Risk of PsA increases steadily for > 60 years following PsO onset. Patient-reported history of PsO characteristics has greater predictive power than physician-measured features at enrollment visits. The characteristics identified in this study provide guidance for screening for PsA risk in patients with PsO.
Collapse
Affiliation(s)
- Sophie Belman
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Jessica A Walsh
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Courtney Carroll
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Michael Milliken
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Benjamin Haaland
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Kristina C Duffin
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Gerald G Krueger
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| | - Bing-Jian Feng
- This study was partially supported by the 2019 Discovery Research Grant and Psoriatic Arthritis Diagnostic Test Grant from the National Psoriasis Foundation, the 2018 Immunology, Inflammation, and Infectious Diseases 3i Initiative at the University of Utah, and Pfizer Inc. (grant numbers WI227108 and WI240276). The support and resources from the Center for High-Performance Computing at the University of Utah are gratefully acknowledged. The computational resources used were partially funded by the National Institutes of Health (NIH) Shared Instrumentation Grant 1S10OD021644-01A1. This project utilized REDCap at the University of Utah, supported by grant 8UL1TR000105 ( formerly UL1RR025764) from the National Center for Advancing Translational Sciences and NIH. S. Belman, MSc, School of Medicine, University of Utah, Salt Lake City, Utah, USA, and Wellcome Sanger Institute, University of Cambridge, Hinxton, UK; J.A. Walsh, MD, C. Carroll, MSc, M. Milliken, MD, MPH, K. Callis Duffin, MD, G.G. Krueger, MD, School of Medicine, University of Utah, Salt Lake City, Utah, USA; B. Haaland, PhD, B.J. Feng, PhD, School of Medicine, and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA. The PERCH software, for which BJF is the inventor, has been nonexclusively licensed to Ambry Genetics Corporation for their clinical genetic testing services and research. BJF also reports funding and sponsorship to his institution on his behalf from Pfizer Inc., Regeneron Genetics Center LLC., and AstraZeneca. BH has consulted for the National Kidney Foundation and Value Analytics Labs. The remaining authors declare no potential conflicts of interest relevant to this article. Address correspondence to Dr. B.J. Feng, 30 N 1900 E, Department of Dermatology, Salt Lake City, UT 84132, USA. . Accepted for publication March 29, 2021
| |
Collapse
|
18
|
Psoriatic arthritis: the role of the nonphysician clinician in the diagnosis and treatment of patients with psoriasis. DRUGS & THERAPY PERSPECTIVES 2021. [DOI: 10.1007/s40267-021-00814-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
19
|
Chandran V, van der Heijde D, Fleischmann RM, Lespessailles E, Helliwell PS, Kameda H, Burgos-Vargas R, Erickson JS, Rathmann SS, Sprabery AT, Birt JA, Shuler CL, Gallo G. Ixekizumab treatment of biologic-naïve patients with active psoriatic arthritis: 3-year results from a phase III clinical trial (SPIRIT-P1). Rheumatology (Oxford) 2021; 59:2774-2784. [PMID: 32031665 PMCID: PMC7516094 DOI: 10.1093/rheumatology/kez684] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/16/2019] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE The aim was to assess the safety and efficacy of up to 156 weeks of ixekizumab (an IL-17A antagonist) treatment in PsA patients. METHODS In a phase III study, patients naïve to biologic treatment were randomized to placebo, adalimumab 40 mg every 2 weeks (ADA; active reference) or ixekizumab 80 mg every 2 weeks (IXEQ2W) or every 4 weeks (IXEQ4W) after an initial dose of 160 mg. At week 24 (week 16 for inadequate responders), ADA (after 8-week washout) and placebo patients were re-randomized to IXEQ2W or IXEQ4W. Outcomes were evaluated using a modified non-responder imputation [linear extrapolation for radiographic progression (modified total Sharp score = 0)] during extended treatment until week 156. RESULTS Of 417 patients, 381 entered the extension, and 243 of 381 (63.8%) completed the 156-week study. Incidence rates of treatment-emergent and serious adverse events, respectively, were 38.0 and 5.2 with IXEQ2W (n = 189) and 38.1 and 8.0 with IXEQ4W (n = 197). One death occurred (IXEQ4W). With IXEQ2W and IXEQ4W, respectively, the response rates persisted to week 156 as measured by the ACR response ≥20% (62.5 and 69.8%), ≥50% (56.1 and 51.8%) and ≥70% (43.8 and 33.4%), psoriasis area and severity index (PASI) 75 (69.1 and 63.5%), PASI 90 (64.5 and 51.2%) and PASI 100 (60.5 and 43.6%). Inhibition of radiographic progression also persisted to week 156 in 61% of IXEQ2W and 71% of IXEQ4W patients. CONCLUSION In this 156-week study of ixekizumab, the safety profile remained consistent with previous reports, and improvements in signs and symptoms of PsA were observed, including persistent low rates of radiographic progression. TRIAL REGISTRATION ClinicalTrials.gov, http://clinicaltrials.gov, NCT01695239, EudraCT 2011-002326-49.
Collapse
Affiliation(s)
- Vinod Chandran
- Division of Rheumatology, Department of Medicine, University of Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Roy M Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric Lespessailles
- Department of Rheumatology CHR Orléans, University of Orléans, Orléans, France
| | | | - Hideto Kameda
- Department of Internal Medicine, Toho University, Tokyo, Japan
| | | | | | | | | | | | | | - Gaia Gallo
- Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
20
|
Serum biomarkers for Modic changes in patients with chronic low back pain. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2021; 30:1018-1027. [PMID: 33423134 DOI: 10.1007/s00586-020-06713-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 11/21/2020] [Accepted: 12/29/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE Lumbar Modic change (MC) can serve as a diagnostic marker as well as an independent source of chronic low back pain (CLBP). This study aimed to test for the existence of serum biomarkers in CLBP patients with MC. METHODS Age- and sex-matched CLBP patients with confirmed MC on lumbar MRI (n = 40) and pain-free controls (n = 40) were assessed. MC was classified into M1, predominating M1, predominating M2 and M2. MC volumes were calculated. Fasting blood samples were assessed for inflammatory mediators, signalling molecules, growth factors and bone turnover markers. Serum concentrations of 46 biomarkers were measured. RESULTS Median concentrations of interleukin (IL)-15 (p < 0.001), IL-8 (p < 0.001), tumour necrosis factor (TNF)-alpha (p < 0.001), Eotaxin-1 (p < 0.05), Eotaxin-3 (p < 0.001), monocyte chemotactic protein (MCP)-1 (p < 0.05), macrophage inflammatory protein (MIP)-1alpha (p < 0.01), TEK receptor tyrosine kinase (Tie)-2 (p < 0.001), vascular cell adhesion molecule (VCAM)-1 (p < 0.001), RANTES (p < 0.001), C telopeptide of type I collagen (CTX)-1 (p < 0.001), vascular endothelial growth factor (VEGF)-C (p < 0.001), VEGF-D (p < 0.05), fms-related tyrosine kinase (Flt)-1 (p < 0.01) and intercellular adhesion molecule (ICAM)-1 (p < 0.01) were significantly higher among controls. IL-1sRII (23.2 vs. 15.5 ng/ml, p < 0.001) and hepatocyte growth factor (HGF)-1 (169 vs. 105 pg/ml, p < 0.01) concentrations were significantly higher among patients. Type or volume of MC was not associated with biomarker concentrations. CONCLUSIONS This is the first study to assess the blood serum biomarker profile in individuals with CLBP with MC. Several biomarkers were suppressed, while two markers (IL-1sRII and HGF) were elevated among MC patients, irrespective of MC type or size, with CLBP compared with asymptomatic controls.
Collapse
|
21
|
Introduction of a Simplified Psoriatic Arthritis Magnetic Resonance Imaging Score (sPsAMRIS): A Potential Tool for Treatment Monitoring in Peripheral Psoriatic Arthritis. Diagnostics (Basel) 2020; 10:diagnostics10121093. [PMID: 33333853 PMCID: PMC7765290 DOI: 10.3390/diagnostics10121093] [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: 11/17/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 12/02/2022] Open
Abstract
Background: To evaluate whether a simplified (s) version of the psoriatic arthritis magnetic resonance imaging score (PsAMRIS), sPsAMRIS, is a potential tool for therapy monitoring in psoriatic arthritis (PsA). Methods: Seventeen patients with active psoriatic arthritis (PsA) underwent magnetic resonance imaging (MRI) at 3 T of the clinically dominant hand at baseline and after 6 months. Scoring was performed by two musculoskeletal radiologists in terms of the PsAMRIS and sPsAMRIS, which is a simplified version with reduced item numbers based on prior evaluation of responsiveness to change by standardized response means (SRMs). Both scores were compared by calculation of overall and each sub-score’s SRMs and relative efficacy (RE) after bootstrapping. Results: PsAMRIS sub-scores of MCP joints 3 and 4, and proximal interphalangeal (PIP) joint 4 had the highest SRM (−0.07 each), indicating highest responsiveness to change, and were, therefore, included in sPsAMRIS. Compared to PsAMRIS, sPsAMRIS was characterized by higher SRMs (sPsAMRIS: −0.13 vs. PsAMRIS: −0.02) and higher RE (29.46). sPsAMRIS and PsAMRIS were highly correlated at baseline (r = 0.75, p < 0.01 (Pearson’s correlation)) and at 6-month follow-up (r = 0.64, p = 0.01). Mean time burden for completion of scoring per MRI study was significantly reduced when using PsAMRIS (469 ± 87.03 s) as compared to sPsAMRIS (140.1 ± 21.25 s) (p < 0.001). Conclusion: Due to its similar responsiveness to change compared to standard PsAMRIS, and time efficiency, sPsAMRIS might be a potential diagnostic tool to quantitatively assess and monitor therapy in PsA.
Collapse
|
22
|
Chen N, Yang H, Li Q, Song L, Gopinath SCB, Wu D. An interdigitated aptasensor to detect interleukin-6 for diagnosing rheumatoid arthritis in serum. Biotechnol Appl Biochem 2020; 68:1479-1485. [PMID: 33244818 DOI: 10.1002/bab.2068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/30/2020] [Indexed: 01/18/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder causing chronic inflammation in the small joints of the articular bone and destruction of articular cartilage. RA causes stiffness, pain, joint destruction, substantial comorbidity, and functional disability. Early-stage diagnosis of RA can help in the treatment of the disease and expand the patient life span. Interleukins are a group of inflammatory cytokines; in particular, an abundance of interleukin-6 (IL-6) was found in the synovial fluid and serum. In RA patients, the levels of IL-6 have been found to be correlated with the disease, and this work focused on detecting IL-6 by its aptamer with the help of a biotin-streptavidin strategy on an interdigitated electrode. A sensitivity of 1 fM (0.021 pg/mL) and a limit of detection of 10 fM (0.21 pg/mL) were found by a linear regression [y = 0.6413x - 0.6249; R² = 0.952] of the linear range from 1 fM to 100 pM. This method enhanced the immobilization of higher aptamer molecules for recognizing RA in serum-containing samples and is applicable to other diseases.
Collapse
Affiliation(s)
- Nan Chen
- Orthopedics Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Hao Yang
- Orthopedics Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Qing Li
- Orthopedics Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Lijun Song
- Geriatric Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Subash C B Gopinath
- Faculty of Chemical Engineering Technology, Universiti Malaysia Perlis (UniMAP), Perlis, Malaysia.,Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, 01000, Malaysia
| | - Di Wu
- Orthopedics Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| |
Collapse
|
23
|
Chimenti MS, D’Antonio A, Conigliaro P, Ferrigno S, Vendola A, Ferraioli M, Triggianese P, Costa L, Caso F, Perricone R. An Update for the Clinician on Biologics for the Treatment of Psoriatic Arthritis. Biologics 2020; 14:53-75. [PMID: 32903867 PMCID: PMC7445514 DOI: 10.2147/btt.s260754] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthropathy typically associated with psoriasis (PsO). The pathogenesis is strictly related to the association among the presence of genetic risk alleles and innate and acquired immune response with dramatic consequences on bone remodeling. Clinically, PsA patients may present heterogenicity of articular and periarticular manifestations that may be associated with the presence of comorbidities making treatment decision challenging in patients management. The identification of patient-targeted therapies is still a critical issue. Actually, several biological and synthetic drugs are promising in terms of efficacy and safety profile. National and international treatment recommendations support clinicians in the decision of the best treatment, although they may have limits basically related to updates and different outcomes included in the clinical studies evaluated. The aim of this narrative review is therefore to give guidance for clinicians for PsA patients treatment. For this purpose, we evaluated evidence on biological therapies efficacy used for PsA treatment. Specifically, we reviewed data on biological therapies, Janus kinases (JAK) inhibitors, and drugs with a new mechanism of action that are part of the treatment pipeline. The concept of "switching" and "swapping" is also described, as well as data concerning special populations such as pregnant women and elderly patients.
Collapse
Affiliation(s)
- Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Arianna D’Antonio
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sara Ferrigno
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Vendola
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Mario Ferraioli
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II, Naples, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
24
|
Garnero P, Landewé R, Chapurlat RD. The role of biochemical markers of joint tissue remodelling to predict progression and treatment efficacy in inflammatory rheumatic diseases. Rheumatology (Oxford) 2020; 59:1207-1217. [PMID: 32011708 DOI: 10.1093/rheumatology/kez647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/02/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Structural damage is a hallmark in RA, spondyloarthropy (SpA) and psoriatric arthritis (PsA). Its progression is difficult to predict and current radiological or inflammatory biological markers lack sensitivity. Biochemical markers of bone, cartilage and synovial tissues provide a dynamic indication of the anabolism and catabolism of joint tissues and can be easily measured by immunoassays. Novel biochemical markers including post-translational modifications of matrix proteins and enzyme-generated neoepitopes with increased tissue and/or biological pathway specificity have been developed. Their evaluation in clinical trials of novel biologic therapies and epidemiological studies indicated that their measurements could be useful to predict progression of structural damage and treatment efficacy, independently of current clinical, radiological and biological indices of disease activity. In this paper we briefly describe the latest developments in biochemical markers and critically analyse the clinical data assessing the utility of established and novel biochemical markers in RA, SpA and PsA.
Collapse
Affiliation(s)
- Patrick Garnero
- INSERM Research Unit 1033-Lyos, Hôpital E. Herriot, Lyon, France
| | - Robert Landewé
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
25
|
Shiraishi M, Fukuda T, Igarashi T, Tokashiki T, Kayama R, Ojiri H. Differentiating Rheumatoid and Psoriatic Arthritis of the Hand: Multimodality Imaging Characteristics. Radiographics 2020; 40:1339-1354. [PMID: 32735474 DOI: 10.1148/rg.2020200029] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Accurate diagnosis and therapeutic intervention at an early stage is paramount for the management of rheumatoid arthritis (RA) and psoriatic arthritis (PsA), which are the two major types of inflammatory arthritis that involve the hand joints. As more disease-specific medications are developed, medication selection according to the correct diagnosis becomes more important. A delay in diagnosis and inappropriate medication selection may result in poor functional prognosis. However, clinical differentiation between RA and PsA can be challenging and may become largely dependent on imaging interpretation results. Although there is substantial overlap in the imaging findings of RA and PsA, there are differences in the affected primary target sites, reflected by the various patterns of joint involvement, and different microanatomic localization of abnormalities within a single joint in each disease. Therefore, appropriate use of various imaging modalities and accurate image interpretation add significant value to the diagnosis and treatment process. The synovio-entheseal complex is an important concept for understanding the imaging features of PsA. The authors review the different features of RA and PsA of the hands seen with various imaging modalities, including radiography, US, MRI, and dual-energy CT, with updates on the contemporary role of imaging in diagnosis and treatment. The radiologist should have sufficient knowledge to interpret imaging findings and understand the strengths and weaknesses of each modality to recommend the appropriate imaging method and differentiate both diseases accurately. ©RSNA, 2020.
Collapse
Affiliation(s)
- Megumi Shiraishi
- From the Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Takeshi Fukuda
- From the Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Takao Igarashi
- From the Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Tadashi Tokashiki
- From the Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Reina Kayama
- From the Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroya Ojiri
- From the Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| |
Collapse
|
26
|
Bai L, Bai Y, Yang Y, Zhang W, Huang L, Ma R, Wang L, Duan H, Wan Q. Baicalin alleviates collagen‑induced arthritis and suppresses TLR2/MYD88/NF‑κB p65 signaling in rats and HFLS‑RAs. Mol Med Rep 2020; 22:2833-2841. [PMID: 32945496 PMCID: PMC7453616 DOI: 10.3892/mmr.2020.11369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/02/2020] [Indexed: 12/19/2022] Open
Abstract
Baicalin is a flavonoid isolated from the root of Scutellaria baicalensis with anti‑inflammatory, antioxidant and antiapoptotic pharmacological properties. however, the therapeutic effect of baicalin on rheumatoid arthritis (RA) is not completely understood. The present study aimed to explore the therapeutic potential and mechanisms underlying baicalin in collagen‑induced arthritis (CIA) model rats. CIA was induced in male SD rats. The hind paw thickness and severity of joint injury were monitored to assess the onset of arthritis. At 28 days after the initial immunization, different doses of baicalin were administered once daily via oral gavage for 40 days. The radiologic and pathological alterations were examined using X‑ray, and hematoxylin and eosin staining, respectively. ELISA was employed to measure the serum levels of proinflammatory cytokines. Reverse transcription‑quantitative PCR and western blotting were conducted to determine the expression of toll‑like receptor (TLR)2, myeloid differentiation factor 88 (MYD88) and NF‑κB p65. Baicalin treatment noticeably alleviated radiographic and histologic abnormalities in the hind paw joints of CIA model rats in a dose‑dependent manner. The serum levels of proinflammatory cytokines were significantly decreased in baicalin‑treated CIA model rats compared with vehicle‑treated CIA model rats. The mRNA expression levels of TLR2 and MYD88, as well as the protein expression levels of TLR2, MYD88 and NF‑κB p65 were significantly decreased by baicalin treatment in the synovial tissue of CIA model rats and human RA fibroblast‑like synoviocytes. The results suggested that baicalin may exert a beneficial effect on CIA, which may be mediated by inhibiting the TLR2/MYD88/NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Lin Bai
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ya Bai
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yuxin Yang
- Department of Clinical Medicine, Medical College, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Wei Zhang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ling Huang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Rui Ma
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Li Wang
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Haizheng Duan
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qiaofeng Wan
- Department of Pathogenic Biology and Immunology, College of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
27
|
Affiliation(s)
- Vinod Chandran
- Faculty of Medicine, University of Toronto, Toronto, Canada
- Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Krembil Research Institute, University Health Network, Toronto, Canada
| |
Collapse
|
28
|
Madav Y, Barve K, Prabhakar B. Current trends in theranostics for rheumatoid arthritis. Eur J Pharm Sci 2020; 145:105240. [DOI: 10.1016/j.ejps.2020.105240] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 01/08/2023]
|
29
|
Krieg L, Schaffert A, Kern M, Landgraf K, Wabitsch M, Beck-Sickinger AG, Körner A, Blüher M, von Bergen M, Schubert K. An MRM-Based Multiplexed Quantification Assay for Human Adipokines and Apolipoproteins. Molecules 2020; 25:molecules25040775. [PMID: 32054032 PMCID: PMC7070386 DOI: 10.3390/molecules25040775] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 12/14/2022] Open
Abstract
Adipokines and apolipoproteins are key regulators and potential biomarkers in obesity and associated diseases and their quantitative assessment is crucial for functional analyses to understand disease mechanisms. Compared to routinely used ELISAs, multiple reaction monitoring (MRM)-based mass spectrometry allows multiplexing and detection of proteins for which antibodies are not available. Thus, we established an MRM method to quantify 9 adipokines and 10 apolipoproteins in human serum. We optimized sample preparation by depleting the two most abundant serum proteins for improved detectability of low abundant proteins. Intra-day and inter-day imprecision were below 16.5%, demonstrating a high accuracy. In 50 serum samples from participants with either normal weight or obesity, we quantified 8 adipokines and 10 apolipoproteins. Significantly different abundances were observed for five adipokines (adipsin, adiponectin, chemerin, leptin, vaspin) and four apolipoproteins (apo-B100/-C2/-C4/-D) between the body mass index (BMI) groups. Additionally, we applied our MRM assay to serum samples from normal weight children and human adipocyte cell culture supernatants to proof the feasibility for large cohort studies and distinct biological matrices. In summary, this multiplexed assay facilitated the investigation of relationships between adipokines or apolipoproteins and phenotypes or clinical parameters in large cohorts, which may contribute to disease prediction approaches in the future.
Collapse
Affiliation(s)
- Laura Krieg
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany; (L.K.)
| | - Alexandra Schaffert
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany; (L.K.)
| | - Matthias Kern
- Department of Medicine, University of Leipzig, Liebigstraße 27b, 04103 Leipzig, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research, Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 20a, 04103 Leipzig, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology Diabetes, Ulm University Medical Center, Eythstraße 24 89075 Ulm, Germany
| | | | - Antje Körner
- Center for Pediatric Research, Hospital for Children & Adolescents, University of Leipzig, Liebigstraße 20a, 04103 Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Liebigstraße 27b, 04103 Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany; (L.K.)
- Institute of Biochemistry, University of Leipzig, Brüderstraße 34, 04103 Leipzig, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, UFZ, Helmholtz-Centre for Environmental Research, Permoserstraße 15, 04318 Leipzig, Germany; (L.K.)
- Correspondence:
| |
Collapse
|
30
|
Pouw J, Leijten E, Radstake T, Boes M. Emerging molecular biomarkers for predicting therapy response in psoriatic arthritis: A review of literature. Clin Immunol 2020; 211:108318. [DOI: 10.1016/j.clim.2019.108318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023]
|
31
|
Kaeley GS, Bakewell C, Deodhar A. The importance of ultrasound in identifying and differentiating patients with early inflammatory arthritis: a narrative review. Arthritis Res Ther 2020; 22:1. [PMID: 31898524 PMCID: PMC6939339 DOI: 10.1186/s13075-019-2050-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/05/2019] [Indexed: 11/13/2022] Open
Abstract
Early differentiation between different types of inflammatory arthritis and subsequent initiation of modern treatments can improve patient outcomes by reducing disease activity and preventing joint damage. Routine clinical evaluation, laboratory testing, and radiographs are typically sufficient for differentiating between inflammatory and predominantly degenerative arthritis (e.g., osteoarthritis). However, in some patients with inflammatory arthritis, these techniques fail to accurately identify the type of early-stage disease. Further evaluation by ultrasound imaging can delineate the inflammatory arthritis phenotype present. Ultrasound is a noninvasive, cost-effective method that enables the evaluation of several joints at the same time, including functional assessments. Further, ultrasound can visualize pathophysiological changes such as synovitis, tenosynovitis, enthesitis, bone erosions, and crystal deposits at a subclinical level, which makes it an effective technique to identify and differentiate most common types of inflammatory arthritis. Limitations associated with ultrasound imaging should be considered for its use in the differentiation and diagnosis of inflammatory arthritides.
Collapse
Affiliation(s)
- Gurjit S Kaeley
- Division of Rheumatology and Clinical Immunology, University of Florida College of Medicine, 653-1 West 8th St., LRC 2nd Floor L-14, Jacksonville, FL, 32209, USA.
| | | | - Atul Deodhar
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
32
|
Diani M, Perego S, Sansoni V, Bertino L, Gomarasca M, Faraldi M, Pigatto PDM, Damiani G, Banfi G, Altomare G, Lombardi G. Differences in Osteoimmunological Biomarkers Predictive of Psoriatic Arthritis among a Large Italian Cohort of Psoriatic Patients. Int J Mol Sci 2019; 20:ijms20225617. [PMID: 31717649 PMCID: PMC6888436 DOI: 10.3390/ijms20225617] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
(1) Background: In literature it is reported that 20–30% of psoriatic patients evolve to psoriatic arthritis over time. Currently, no specific biochemical markers can either predict progression to psoriatic arthritis or response to therapies. This study aimed to identify osteoimmunological markers applicable to clinical practice, giving a quantitative tool for evaluating pathological status and, eventually, to provide prognostic support in diagnosis. (2) Methods: Soluble (serum) bone and cartilage markers were quantified in 50 patients with only psoriasis, 50 psoriatic patients with psoriatic arthritis, and 20 healthy controls by means of multiplex and enzyme-linked immunoassays. (3) Results: Differences in the concentrations of matrix metalloproteases (MMPs), tissue inhibitors of metalloproteinases (TIMPs), receptor activator of nuclear factor kappa-B- ligand (RANK-L), procollagen type I N propeptide (PINP), C-terminal telopeptide of type I collagen (CTx-I), dickkopf-related protein 1 (DKK1), and sclerostin (SOST) distinguished healthy controls from psoriasis and psoriatic arthritis patients. We found that MMP2, MMP12, MMP13, TIMP2, and TIMP4 distinguished psoriasis from psoriatic arthritis patients undergoing a systemic treatment, with a good diagnostic accuracy (Area under the ROC Curve (AUC) > 0.7). Then, chitinase-3-like protein 1 (CHI3L1) and MMP10 distinguished psoriasis from psoriatic arthritis not undergoing systemic therapy and, in the presence of onychopathy, MMP8 levels were higher in psoriasis than in psoriatic arthritis. However, in these latter cases, the diagnostic accuracy of the identified biomarkers was low (0.5 < AUC < 0.7). (4) Conclusions. By highlighting never exploited differences, the wide osteoimmunological biomarkers panel provides a novel clue to the development of diagnostic paths in psoriasis and psoriasis-associated arthropathic disease.
Collapse
Affiliation(s)
- Marco Diani
- Department of Dermatology and Venereology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (M.D.); (P.D.M.P.); (G.A.)
| | - Silvia Perego
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); (V.S.); (M.G.); (M.F.); (G.B.); (G.L.)
| | - Veronica Sansoni
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); (V.S.); (M.G.); (M.F.); (G.B.); (G.L.)
| | - Lucrezia Bertino
- Department of Clinical and Experimental Medicine, section of Dermatology, University of Messina, 98122 Messina, Italy;
| | - Marta Gomarasca
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); (V.S.); (M.G.); (M.F.); (G.B.); (G.L.)
| | - Martina Faraldi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); (V.S.); (M.G.); (M.F.); (G.B.); (G.L.)
| | - Paolo Daniele Maria Pigatto
- Department of Dermatology and Venereology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (M.D.); (P.D.M.P.); (G.A.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milano, Italy
| | - Giovanni Damiani
- Department of Dermatology and Venereology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (M.D.); (P.D.M.P.); (G.A.)
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milano, Italy
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
- Young Dermatologists Italian Network, Centro Studi GISED, 24121 Bergamo, Italy
- Correspondence: ; Tel.: +39-0266214068
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); (V.S.); (M.G.); (M.F.); (G.B.); (G.L.)
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Gianfranco Altomare
- Department of Dermatology and Venereology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (M.D.); (P.D.M.P.); (G.A.)
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); (V.S.); (M.G.); (M.F.); (G.B.); (G.L.)
- Department of Physiology and Pharmacology, Gdańsk University of Physical Education and Sport, 80336 Gdańsk, Poland
| |
Collapse
|
33
|
Abstract
Accurate diagnosis of inflammatory arthritides remains a challenge because of substantial clinical overlap. To achieve a granular classification for informing clinical decisions, numerous potential serologic biomarkers have been identified. Rheumatologists have settled on rheumatoid factor and anti-citrullinated protein antibodies for the diagnosis of rheumatoid arthritis (RA) based on specificity and sensitivity and their ability to be integrated into clinical algorithms. These biomarkers should be interpreted in their specific clinical context. This article discusses the serologic basis for the diagnosis of RA, how these biomarkers have framed conceptualization of the pathogenesis of RA, and the inherent limitations in their use.
Collapse
|
34
|
Implication of IL-17 in Bone Loss and Structural Damage in Inflammatory Rheumatic Diseases. Mediators Inflamm 2019; 2019:8659302. [PMID: 31485194 PMCID: PMC6710740 DOI: 10.1155/2019/8659302] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/29/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
Proinflammatory cytokines play an important role in the systemic and focal bone loss associated with chronic inflammatory diseases. Targeting these cytokines with biologics and small molecules has led to a major improvement of the bone health of patients with inflammatory arthritis. Cytokines from the IL-17 family have been shown to be involved in the pathogenesis of several diseases such as spondyloarthritis, psoriatic arthritis, or psoriasis. IL-17A has been the first described and the most studied. The recent development of targeted therapies against IL-17A or its receptor and their efficacy has confirmed the importance of this cytokine in the development of inflammatory diseases. The aim of this review was to describe the effects of the IL-17 family and more particularly of IL-17A on bone and cartilage tissues. At the cellular level, IL-17A is proosteoclastogenic whereas its effects on osteoblasts depend on the stage of differentiation of these cells. In vivo, IL-17A is not required for normal bone homeostasis but plays an important role in bone loss notably in an ovariectomized mouse model of osteoporosis. Preliminary data from clinical trials showed a stabilisation of bone density in patients treated with anti-IL-17A antibodies. IL-17A plays a central role in the cartilage damage through the induction of collagenases and by decreasing the expression of their inhibitors in synergy with the other proinflammatory cytokines. The prevention of structural damage by anti-IL-17A therapies has been demonstrated in several pivotal clinical trials. Overall, blocking the IL-17A pathway seems to have a positive effect on the bone and cartilage damage observed in inflammatory arthritis. Differences and specificity of these effects compared to those already described with other biologics such as anti-TNF therapies remain to be explored.
Collapse
|
35
|
van der Heijde D, Gladman DD, FitzGerald O, Kavanaugh A, Graham D, Wang C, Fallon L. Radiographic Progression According to Baseline C-reactive Protein Levels and Other Risk Factors in Psoriatic Arthritis Treated with Tofacitinib or Adalimumab. J Rheumatol 2019; 46:1089-1096. [DOI: 10.3899/jrheum.180971] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2018] [Indexed: 01/30/2023]
Abstract
Objective.To evaluate the effect of baseline risk factors on radiographic progression in patients with active psoriatic arthritis (PsA) who had an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (csDMARD) and were treated with tofacitinib or adalimumab (ADA).Methods.Tofacitinib is an oral Janus kinase inhibitor for the treatment of PsA. OPAL Broaden was a 12-month, double-blind phase III trial. Patients received tofacitinib 5 mg twice daily (BID; n = 107), tofacitinib 10 mg BID (n = 104), or ADA 40 mg once every 2 weeks (n = 106), all with 1 background csDMARD. Radiographs (baseline and Month 12) were scored using the van der Heijde-modified total Sharp score (mTSS) for PsA. Radiographic nonprogression was defined as an increase from baseline in mTSS ≤ 0.5, ≤ 0, or ≤ 0.66. Changes from baseline in mTSS and nonprogression (≤ 0.5 increase from baseline in mTSS) were analyzed by baseline C-reactive protein (CRP) > 2.87 or ≤ 2.87 mg/l. Baseline predictors of radiographic progression were analyzed.Results.At Month 12, > 90% of patients receiving tofacitinib or ADA met all radiographic nonprogression criteria. Mean changes from baseline through Month 12 in mTSS, erosion, and joint space narrowing scores were close to 0. Changes in radiographic outcomes were minimal, irrespective of baseline CRP levels > 2.87 or ≤ 2.87 mg/l, with a small numerical difference observed for tofacitinib 5 mg BID. A significant relationship was observed between baseline CRP level and increases from baseline in mTSS > 0.5 at Month 12.Conclusion.Elevated CRP levels at baseline were associated with greater structural progression. Changes in radiographic outcomes were minimal regardless of CRP levels. [Clinical trial registration number (www.ClinicalTrials.gov): NCT01877668]
Collapse
|
36
|
Zhang R, Yang X, Wang J, Han L, Yang A, Zhang J, Zhang D, Li B, Li Z, Xiong Y. Identification of potential biomarkers for differential diagnosis between rheumatoid arthritis and osteoarthritis via integrative genome‑wide gene expression profiling analysis. Mol Med Rep 2018; 19:30-40. [PMID: 30483789 PMCID: PMC6297798 DOI: 10.3892/mmr.2018.9677] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/24/2018] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to identify potential novel biomarkers in synovial tissue obtained from patients with Rheumatoid Arthritis (RA) and Osteoarthritis (OA) for differential diagnosis. The genome-wide expression profiling datasets of synovial tissues from RA and OA cohorts, including GSE55235, GSE55457 and GSE55584 datasets, were retrieved and used to identify differentially expressed genes (DEGs; P<0.05; false discovery rate <0.05 and Fold Change >2) between RA and OA using R software. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of DEGs were performed to determine molecular and biochemical pathways associated with the identified DEGs, and a protein-protein interaction (PPI) network of the DEGs was constructed using Cytoscape software. Significant modules in the PPI network and candidate driver genes were screened using the Molecular Complex Detection Algorithm. Potential biomarkers were evaluated by receiver operating characteristic and logistic regression analyses. Large numbers of DEGs were detected, including 273, 205 and 179 DEGs in the GSE55235, GSE55457 and GSE55584 datasets, respectively. Among them, 80 DEGs exhibited identical expression trends in all the three datasets, including 49 upregulated and 31 downregulated genes in patients with RA. DEGs in patients suffering from RA compared with patients suffering from OA were predominantly associated with the primary immunodeficiency pathway, including interleukin 7 receptor (IL7R) and signal transducer activator of transcription 1 (STAT1). The sensitivity of IL7R + STAT1 to differentiate RA from OA was 93.94% with a specificity of 80.77%. The results generated from analyses of the GSE36700 dataset were closely associated with results generated from analyses of GSE55235, GSE55457 and GSE55584 datasets, which further verified the reliability of the aforementioned results. The results of the present study suggested that increased expression of IL7R and STAT1 in synovial tissue as well as in the primary immunodeficiency may be associated with RA occurrence. These identified novel biomarkers may be used to predict disease occurrence and clinically differentiate RA from OA.
Collapse
Affiliation(s)
- Rongqiang Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoli Yang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Wang
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, P.R. China
| | - Lixin Han
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| | - Aimin Yang
- School of Public Health, Brown University, Providence, RI 02906, USA
| | - Jie Zhang
- School of Public Health, Brown University, Providence, RI 02906, USA
| | - Dandan Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| | - Baorong Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| | - Zhaofang Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| | - Yongmin Xiong
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases of The National Health and Family Planning Commission, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
37
|
Ha YJ, Han DW, Kim JH, Chung SW, Kang EH, Song YW, Lee YJ. Circulating Semaphorin 4D as a Marker for Predicting Radiographic Progression in Patients with Rheumatoid Arthritis. DISEASE MARKERS 2018; 2018:2318386. [PMID: 30538782 PMCID: PMC6261241 DOI: 10.1155/2018/2318386] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
Abstract
Semaphorin 3A (Sema3A) and semaphorin 4D (Sema4D) are molecules which regulate immune responses as well as bone remodeling process. The aim of this study was to evaluate the serum levels of Sema3A and Sema4D and to investigate their clinical significance in rheumatoid arthritis (RA). The serum levels of Sema3A and Sema4D were measured in 130 patients with RA and 65 sex- and age-matched healthy individuals. Circulating levels of biomarkers of RA-related inflammation and bone turnover such as tumor necrosis factor- (TNF-) α, interleukin- (IL-) 6, IL-22, IL-34, osteopontin, Dkk-1, and sclerostin were also measured. Disease activity was determined by the 28-joint disease activity score (DAS28), and radiographic joint damage was assessed by the modified Sharp van der Heijde score (SHS). The serum levels of Sema3A were significantly higher in patients with RA than those in healthy controls (p < 0.001), whereas serum4D levels did not differ between the two groups. The levels of Sema4D showed a positive correlation with C-reactive protein (p = 0.001) and IL-6 (p < 0.001) levels, whereas the levels of Sema3A showed a negative correlation with Dkk-1 (p = 0.007) and TNF-α (p = 0.001). Even though Sema3A and Sema4D levels were comparable between RA patients with DAS28> 3.2 and with DAS28 ≤ 3.2, RA patients with radiographic progression (ΔSHS change/year ≥ 1) had significantly higher baseline levels of Sema4D than those without progression (p = 0.029). Additionally, when RA patients were divided into 3 groups using tertiles of Sema4D levels, the percentage of progressors was significantly increased (p = 0.045). In multivariate logistic regression analysis, serum Sema4D levels were an independent risk factor for radiographic progression. Our results suggest that the baseline levels of Sema4D might be a useful marker to identify RA patients with subsequent radiographic progression and that Sema4D may be an active mediator involved in RA-induced joint damage.
Collapse
Affiliation(s)
- You-Jung Ha
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dong Woo Han
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Hyoun Kim
- Division of Rheumatology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Sang Wan Chung
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Eun Ha Kang
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yeong Wook Song
- WCU Department of Molecular Medicine and Biopharmaceutical Sciences, Medical Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yun Jong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Translational Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
38
|
Abraham S, Barton A, Eder L, Lim A, McGonagle D, McHugh N, Pennington S, Sengupta R, Siebert S, Bowness P, Schafer PH, Cullen E, FitzGerald O. Advancing research paradigms and pathophysiological pathways in psoriatic arthritis and ankylosing spondylitis: Proceedings of the 2017 Platform for the Exchange of Expertise and Research (PEER) meeting. Semin Arthritis Rheum 2018; 48:1005-1013. [PMID: 30415944 DOI: 10.1016/j.semarthrit.2018.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/06/2018] [Accepted: 10/09/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Sonya Abraham
- NIHR/Wellcome Trust Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College Healthcare NHS Trust, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK.
| | - Anne Barton
- Centre for Musculoskeletal Research, University of Manchester and Central Manchester NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Lihi Eder
- Women's College Research Institute, Women's College Hospital, University of Toronto, Toronto, ON, Canada
| | - Adrian Lim
- Charing Cross Hospital, Imperial College London, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Neil McHugh
- Royal National Hospital for Rheumatic Diseases and Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Stephen Pennington
- St. Vincent's University Hospital and Conway Institute for Biomolecular Research, University College Dublin School of Medicine, Dublin, Ireland
| | - Raj Sengupta
- Royal National Hospital for Rheumatic Diseases, Bath, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Paul Bowness
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Science, Oxford OX3 7LD, UK
| | | | | | - Oliver FitzGerald
- St. Vincent's University Hospital and Conway Institute for Biomolecular Research, University College Dublin School of Medicine, Dublin, Ireland
| |
Collapse
|
39
|
Role of cartilage oligomeric matrix protein (COMP) as a prognostic biomarker in follow-up of early rheumatoid arthritis patients: Correlation to musculoskeletal ultrasonographic findings. EGYPTIAN RHEUMATOLOGIST 2018. [DOI: 10.1016/j.ejr.2018.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
40
|
Merola JF, Espinoza LR, Fleischmann R. Distinguishing rheumatoid arthritis from psoriatic arthritis. RMD Open 2018; 4:e000656. [PMID: 30167326 PMCID: PMC6109814 DOI: 10.1136/rmdopen-2018-000656] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/25/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022] Open
Abstract
Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) have key differences in clinical presentation, radiographic findings, comorbidities and pathogenesis to distinguish between these common forms of chronic inflammatory arthritis. Joint involvement is typically, but not always, asymmetric in PsA, while it is predominantly symmetric in RA. Bone erosions, without new bone growth, and cervical spine involvement are distinctive of RA, while axial spine involvement, psoriasis and nail dystrophy are distinctive of PsA. Patients with PsA typically have seronegative test findings for rheumatoid factor (RF) and cyclic citrullinated peptide (CCP) antibodies, while approximately 80% of patients with RA have positive findings for RF and CCP antibodies. Although there is overlap in the pathogenesis of PsA and RA, differences are also present that affect the efficacy of treatment. In PsA, levels of interleukin (IL)-1β, IL-6, IL-17, IL-22, IL-23, interferon-γ and tumour necrosis factor-α (TNF-α) are elevated, and in RA, levels of IL-1, IL-6, IL-22, IL-33, TNF-α, chemokine ligand 11 and chemokine C-X-C motif ligand 13 are elevated. Differences in the pathogenesis of RA and PsA translate into some variances in the specificity and efficacy of therapies.
Collapse
Affiliation(s)
- Joseph F Merola
- Department of Dermatology, Medicine and Rheumatology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Luis R Espinoza
- Section of Rheumatology, LSU Health Sciences Center at New Orleans, New Orleans, Louisiana, USA
| | - Roy Fleischmann
- Department of Medicine, University of Texas Southwestern Medical Center, Metroplex Clinical Research Center, Dallas, Texas, USA
| |
Collapse
|
41
|
Llibre A, Duffy D. Immune response biomarkers in human and veterinary research. Comp Immunol Microbiol Infect Dis 2018; 59:57-62. [PMID: 30290889 PMCID: PMC7172169 DOI: 10.1016/j.cimid.2018.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/22/2018] [Accepted: 09/17/2018] [Indexed: 11/30/2022]
Abstract
Biomarkers are increasingly utilised in biological research and clinical practice for diagnosis of disease, monitoring of therapeutic prognosis, or as end points in clinical studies. Cytokines are small molecules that orchestrate immune responses and as such have great potential as biomarkers for both human and veterinary fields. Given the ease of sampling in the blood, and their high prevalence in clinical applications we will focus on protein detection as an area for biomarker discovery. This is facilitated by new technological developments such as digital ELISA that have led to significant increases in sensitivity. Two highly relevant examples include type I interferons, namely IFNα, that is now directly quantifiable by digital ELISA from biological samples. The application of this approach to the study of the unique bat interferon response may reveal novel findings with applications in both human and veterinary research. As a second example we will describe the use of CXCL10 as a disease biomarker in Tuberculosis, highlighting findings from human and mouse studies that should be considered in veterinary research. In summary, we describe how cytokines may be applied as novel biomarkers and illustrate two key examples where human and veterinary research may complement each other in line with the One Health objectives.
Collapse
Affiliation(s)
- Alba Llibre
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France; INSERM U1223, Paris, France
| | - Darragh Duffy
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France; INSERM U1223, Paris, France.
| |
Collapse
|
42
|
Kessel C, McArdle A, Verweyen E, Weinhage T, Wittkowski H, Pennington SR, Foell D. Proteomics in Chronic Arthritis-Will We Finally Have Useful Biomarkers? Curr Rheumatol Rep 2018; 20:53. [PMID: 30008153 DOI: 10.1007/s11926-018-0762-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Current technical advances enable the assessment of the complex changes in body fluid proteomes and thus allow for the discovery of biomarker signatures rather than just following differences of a single marker. In this review, we aim to summarize current approaches to discover and evaluate multi-biomarker panels for improved monitoring of chronic arthritis disease activity. RECENT FINDINGS Mass spectrometry and affinity proteomic methodologies have been used to identify biomarker panels in synovial fluid, serum, plasma, or urine of pediatric and adult chronic arthritis patients. Notably, despite the numerous efforts to develop new and better biomarker panels, very few have undergone extensive analytical and clinical validation and been adopted into routine use for patient benefit. There remains a significant gap between discovery of chronic arthritis biomarker signatures and their validation for clinical use.
Collapse
Affiliation(s)
- Christoph Kessel
- Department of Paediatric Rheumatology and Immunology, University of Muenster, Domagkstraße 3, 48149, Muenster, Germany
| | - Angela McArdle
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Emely Verweyen
- Department of Paediatric Rheumatology and Immunology, University of Muenster, Domagkstraße 3, 48149, Muenster, Germany
| | - Toni Weinhage
- Department of Paediatric Rheumatology and Immunology, University of Muenster, Domagkstraße 3, 48149, Muenster, Germany
| | - Helmut Wittkowski
- Department of Paediatric Rheumatology and Immunology, University of Muenster, Domagkstraße 3, 48149, Muenster, Germany
| | - Stephen R Pennington
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Dirk Foell
- Department of Paediatric Rheumatology and Immunology, University of Muenster, Domagkstraße 3, 48149, Muenster, Germany.
| |
Collapse
|
43
|
Ghozlani I, Mounach A, Ghazi M, Kherrab A, Niamane R, El Maghraoui A. Influence of anti-cyclic citrullinated peptide on disease activity, structural severity, and bone loss in Moroccan women with rheumatoid arthritis. THE EGYPTIAN RHEUMATOLOGIST 2018. [DOI: 10.1016/j.ejr.2017.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
Mahendran SM, Chandran V. Exploring the Psoriatic Arthritis Proteome in Search of Novel Biomarkers. Proteomes 2018; 6:proteomes6010005. [PMID: 29364831 PMCID: PMC5874764 DOI: 10.3390/proteomes6010005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/16/2018] [Accepted: 01/21/2018] [Indexed: 12/28/2022] Open
Abstract
Psoriatic arthritis (PsA) is an inflammatory arthritis which develops in up to one-third of patients suffering from the cutaneous disorder, psoriasis. The complex and heterogeneous nature of PsA renders it difficult to diagnose, leading to poor outcomes and, therefore, warrants an examination into soluble biomarkers, which may facilitate early detection of the disease. Protein biomarkers are a dynamic resource of pathophysiological information able to provide an immediate reflection of pathological changes caused by disease. Investigations of the serum and synovial fluid of PsA patients has provided new insights into the molecular basis of this disease and led to the identification of sensitive diagnostic and prognostic biomarkers. The collection of novel PsA biomarkers identified through proteomic studies has been reviewed below.
Collapse
Affiliation(s)
- Shalini M Mahendran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada.
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada.
| | - Vinod Chandran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada.
- Centre for Prognosis Studies in Rheumatic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON M5T 1M8, Canada.
- Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, ON M5T 1A1, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON M5T 1A1, Canada.
| |
Collapse
|
45
|
Goulooze SC, Krekels EH, van Dijk M, Tibboel D, van der Graaf PH, Hankemeier T, Knibbe CA, van Hasselt JC. Towards personalized treatment of pain using a quantitative systems pharmacology approach. Eur J Pharm Sci 2017; 109S:S32-S38. [DOI: 10.1016/j.ejps.2017.05.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 02/08/2023]
|
46
|
Marton N, Kovács OT, Baricza E, Kittel Á, Győri D, Mócsai A, Meier FMP, Goodyear CS, McInnes IB, Buzás EI, Nagy G. Extracellular vesicles regulate the human osteoclastogenesis: divergent roles in discrete inflammatory arthropathies. Cell Mol Life Sci 2017; 74:3599-3611. [PMID: 28493076 PMCID: PMC11107760 DOI: 10.1007/s00018-017-2535-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 03/26/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Extracellular vesicles (EVs) are subcellular signalosomes. Although characteristic EV production is associated with numerous physiological and pathological conditions, the effect of blood-derived EVs on bone homeostasis is unknown. Herein we evaluated the role of circulating EVs on human osteoclastogenesis. METHODS Blood samples from healthy volunteers, rheumatoid arthritis (RA) and psoriatic arthritis (PsA) patients were collected. Size-based EV sub-fractions were isolated by gravity-driven filtration and differential centrifugation. To investigate the properties of EV samples, resistive pulse sensing technique, transmission electron microscopy, flow cytometry and western blot were performed. CD14+ monocytes were separated from PBMCs, and stimulated with recombinant human M-CSF, RANKL and blood-derived EV sub-fractions. After 7 days, the cells were fixed and stained for tartrate-resistant acid phosphatase and counted. RESULTS EVs isolated by size-based sub-fractions were characterized as either microvesicles or exosomes (EXO). Healthy (n = 11) and RA-derived (n = 12) EXOs profoundly inhibited osteoclast differentiation (70%, p < 0.01; 65%, p < 0.01, respectively). In contrast, PsA-derived (n = 10) EXOs had a stimulatory effect (75%, p < 0.05). In cross-treatment experiments where EXOs and CD14+ cells were interchanged between the three groups, only healthy (n = 5) and RA (n = 5)-derived EXOs inhibited (p < 0.01, respectively) the generation of osteoclasts in all groups, whereas PsA (n = 7)-derived EXOs were unable to mediate this effect. CONCLUSIONS Our data suggest that blood-derived EXOs are novel regulators of the human osteoclastogenesis and may offer discrete effector function in distinct inflammatory arthropathies.
Collapse
Affiliation(s)
- Nikolett Marton
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Orsolya Tünde Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Eszter Baricza
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - Ágnes Kittel
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Dávid Győri
- Department of Physiology, Semmelweis University, Budapest, Hungary
- MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University, Budapest, Hungary
- MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences and the Semmelweis University, Budapest, Hungary
| | - Florian M P Meier
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary
| | - György Nagy
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, 1089, Budapest, Hungary.
- Department of Rheumatology, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
47
|
Wang F, Wan J, Li Q, Zhang M, Wan Q, Ji C, Li H, Liu R, Han M. Lysyl oxidase is involved in synovial hyperplasia and angiogenesis in rats with collagen‑induced arthritis. Mol Med Rep 2017; 16:6736-6742. [PMID: 28901438 PMCID: PMC5865828 DOI: 10.3892/mmr.2017.7436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 06/13/2017] [Indexed: 12/29/2022] Open
Abstract
Lysyl oxidase (LOX) serves an important role in remodeling the extracellular matrix and angiogenesis in various types of cancer; however, whether LOX is involved in the pathogenesis of rheumatoid arthritis remains unknown. In order to investigate this in the present study, β-aminopropionitrile, an inhibitor of LOX, was injected intraperitoneally into rats with type II collagen-induced arthritis (CIA). Subsequently, synovial hyperplasia was examined by hematoxyl in and eosin staining, and the microvascular density (MVD) and expression levels of LOX, matrix metalloproteinase (MMP)-2 and MMP-9 in the synovial membrane and fluid were determined by immunohistochemistry and ELISA, respectively. The enzyme activity of LOX was evaluated by the Amplex Red Hydrogen Peroxide method. The results demonstrated an increased amount of rough synovial membranes, higher MVD in these membranes and more synovial cell layers in CIA rats compared with in the control rats. In addition, higher enzymatic activity of LOX and higher expression levels of MMP-2 and MMP-9 were revealed in CIA rats compared with in the control rats. Notably, β-aminopropionitrile inhibited paw swelling and the decreased the arthritis index, the MVD in the synovial membranes and the expression levels of MMP-2 and MMP-9. Furthermore, the expression level of LOX in the synovial membranes was positively associated with the MVD and the expression levels of MMP-2 and MMP-9, suggesting that LOX promotes synovial hyperplasia and angiogenesis and that LOX may be a potential therapeutic target for rheumatoid arthritis.
Collapse
Affiliation(s)
- Fan Wang
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Juan Wan
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Qiuyan Li
- Department of Pathogenic Biology and Immunology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Mingzhu Zhang
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Qiaofeng Wan
- Department of Pathogenic Biology and Immunology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Chen Ji
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Haibo Li
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Rongqing Liu
- Department of Rheumatology and Immunology, The General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China
| | - Mei Han
- Department of Pathogenic Biology and Immunology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
48
|
Park YJ, Cho YR, Oh JS, Ahn EK. Effects of Tribulus terrestris on monosodium iodoacetate‑induced osteoarthritis pain in rats. Mol Med Rep 2017; 16:5303-5311. [PMID: 28849084 PMCID: PMC5647062 DOI: 10.3892/mmr.2017.7296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Tribulus terrestris L. (T. terrestris) has been used as a traditional medicine for the treatment of diuretic, lithontriptic, edema and urinary infections. Previous studies have indicated that it is effective in improving inflammation by regulating tumor necrosis factor‑α (TNF)‑α, interleukin (IL)‑6, IL‑10, nitric oxide (NO) and cyclooxygenase (COX)‑2. However, the effects and mechanism of action of T. terrestris on osteoarthritis (OA) remain unknown. Therefore, the present study aimed to evaluate the effects of the ethanolic extract of T. terrestris (ETT) in a monosodium iodoacetate (MIA)‑induced OA animal model. OA was induced in LEW/SSNHSD rats by intra‑articular injection of MIA. Morphometric changes and parameters of the tibial trabecular bone were determined using micro‑computed tomography. The molecular mechanisms of ETT in OA were investigated using reverse transcription‑polymerase chain reaction, western blotting and gelatin zymogram analysis. Treatment with ETT attenuated MIA‑induced OA, and this effect was mediated by the downregulation of NO synthase 2, COX‑2, TNF‑α and IL‑6. Furthermore, the ETT‑mediated attenuation of OA was also dependent on the expression of matrix metalloproteinases‑2 and ‑9. The results of the current study indicate that further evaluation of the mechanisms underlying the attenuation of MIA‑induced OA by ETT are required, and may support the development of ETT as a potential therapeutic agent for the treatment of inflammatory diseases such as OA.
Collapse
Affiliation(s)
- Young Jin Park
- Bio‑center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi 443‑270, Republic of Korea
| | - Young-Rak Cho
- Bio‑center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi 443‑270, Republic of Korea
| | - Joa Sub Oh
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam 330‑714, Republic of Korea
| | - Eun-Kyung Ahn
- Bio‑center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi 443‑270, Republic of Korea
| |
Collapse
|
49
|
Clinical Features of Psoriatic Arthritis: a Comprehensive Review of Unmet Clinical Needs. Clin Rev Allergy Immunol 2017; 55:271-294. [DOI: 10.1007/s12016-017-8630-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
50
|
Seo JY, Suh CH, Jung JY, Kim AR, Yang JW, Kim HA. The neutrophil-to-lymphocyte ratio could be a good diagnostic marker and predictor of relapse in patients with adult-onset Still's disease: A STROBE-compliant retrospective observational analysis. Medicine (Baltimore) 2017; 96:e7546. [PMID: 28723775 PMCID: PMC5521915 DOI: 10.1097/md.0000000000007546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) is the proportion of absolute neutrophil count to lymphocytes on routine complete blood count (CBC) tests, and has been studied as a simple marker of the systemic inflammatory response. This study was performed to investigate whether the NLR could be used as a tool to diagnose and predict prognosis in cases of adult-onset Still's disease (AOSD).We retrospectively reviewed 164 patients with suspected AOSD. Among 164 patients with suspected AOSD, 37 patients received another diagnosis (such as viral infection) and were compared with the 127 patients who received a diagnosis of AOSD. Laboratory tests including CBCs, ferritin, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP) level, and the NLR were evaluated.AOSD patients showed higher neutrophil counts, lower lymphocyte counts, higher NLRs, and higher levels of ferritin, ESR, and CRP than non-AOSD patients (all P < .001). In receiver operating characteristic (ROC) curve analysis of the NLR for diagnosis of AOSD, the area under the curve (AUC) was highest at 0.967 (95% CI = 0.940-0.993) with a cutoff value of 3.08. The cutoff value showed the greatest sensitivity (91.7%), specificity (68.4%), and AUC value (0.967) as a diagnostic tool for AOSD. The NLR and treatment appeared to be significant prognostic factors for relapse, but only age showed a significant relationship with death. Furthermore, the NLR was significantly higher in patients with macrophage activation syndrome than in hemophagocytic lymphohistiocytosis (HLH) patients (P = .007). In ROC analysis, the NLR with a cutoff value of 5.86 showed a sensitivity of 89.4%, specificity of 87.8%, and AUC of 0.794.The NLR can be used as a diagnostic tool and predictor of relapse in AOSD, and for differential diagnosis of HLH.
Collapse
|