1
|
Yang A, Lin L, Zhang J, Wu Y, Zhao Z. A novel role for endoplasmic reticulum protein ERp72 in the pathogenesis of autoantibody-induced arthritis. Scand J Rheumatol 2025; 54:16-24. [PMID: 38975658 DOI: 10.1080/03009742.2024.2362040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/28/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE The family of protein disulphide isomerases (PDIs) is a group of oxidoreductases that catalyze the oxidation, reduction and isomerization of disulphide bonds. Recent studies have shown that overexpression of one of the family enzymes, ERp46, potentiates arthritis severity, suggesting that the PDI family participates in arthritis pathogenesis. This study investigated the role of another PDI member, ERp72, in autoantibody-induced arthritis. METHODS Using the Cre-LoxP method, a mouse strain lacking ERp72 (ERp72-/- mice) was generated. Autoantibody-induced arthritis was induced in both ERp72-/- and ERp72+/+ control mice by injecting serum from K/BxN mice. The synovial inflammation severity was evaluated by joint diameter measurements and histological analysis. Proinflammatory cytokines expression in joint tissue and plasma was assessed by quantitative real-time PCR and ELISA. RESULTS : The absence of ERp72 in the joints, white blood cells, spleen, thymus, and bone marrow of ERp72-/- mice was confirmed. In the K/BxN serum transfer-induced arthritis (STIA) model, ERp72-/- mice exhibited exacerbated arthritis compared to ERp72+/+ mice, with greater joint swelling, bone and cartilage erosion, and synovial inflammation. Furthermore, ERp72-/- mice exhibited increased expression of IL-1β, IL-6 and TNF-α in inflamed joint tissues and higher IL-6 levels in plasma. Conversely, IL-10 levels were lower in ERp72-/- mice inflamed joints than in ERp72+/+ mice. Notably, the basal TNF-α level in the blood of ERp72-/- mice was significantly higher than in ERp72+/+ mice. CONCLUSION ERp72 plays a key role in the negative regulation of autoantibody-induced arthritis.
Collapse
Affiliation(s)
- A Yang
- Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, PR China
| | - L Lin
- Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, PR China
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, PR China
| | - J Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Hematology, Shijiazhuang, PR China
| | - Y Wu
- Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, PR China
| | - Z Zhao
- Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, PR China
| |
Collapse
|
2
|
Kumar A, Ashawat MS, Pandit V, Kumar P. Bioelectronic Medicines-A Novel Approach of Therapeutics in Current Epoch. Curr Pharm Des 2025; 31:163-178. [PMID: 39313906 DOI: 10.2174/0113816128326489240827100537] [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: 04/27/2024] [Accepted: 07/19/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Bioelectronic medicines aim to diagnose and treat a wide range of illnesses and ailments, including cancer, rheumatoid arthritis, inflammatory bowel disease, obesity, diabetes, asthma, paralysis, blindness, bleeding, ischemia, organ transplantation, cardiovascular disease, and neurodegenerative diseases. The focus of bioelectronic medicine is on electrical signaling of the nervous system. Understanding the nervous system's regulatory roles and developing technologies that record, activate, or inhibit neural signaling to influence particular biological pathways. OBJECTIVE Bioelectronic medicine is an emerging therapeutic option with the interconnection between molecular medicine, neuroscience, and bioengineering. The creation of nerve stimulating devices that communicate with both the central and peripheral nervous systems has the potential to completely transform how we treat disorders. Although early clinical applications have been largely effective across entire nerves, the ultimate goal is to create implantable, miniature closed-loop systems that can precisely identify and modulate individual nerve fibers to treat a wide range of disorders. METHODOLOGY The data bases such as PubMed, and Clinicaltrial.gov.in were searched for scientific research, review and clinical trials on bioelectronic medicine. CONCLUSION The field of bioelectronic medicine is trending at present. In recent years, researchers have extended the field's applications, undertaken promising clinical trials, and begun delivering therapies to patients, thus creating the groundwork for significant future advancements. Countries and organizations must collaborate across industries and regions to establish an atmosphere and guidelines that foster the advancement of the field and the fulfillment of its prospective advantages.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Pharmaceutics, Laureate Institute of Pharmacy, Kathog, Jwalamukhi, H.P., India
| | - Mahendra Singh Ashawat
- Department of Pharmaceutics, Laureate Institute of Pharmacy, Kathog, Jwalamukhi, H.P., India
| | - Vinay Pandit
- Department of Pharmaceutics, Laureate Institute of Pharmacy, Kathog, Jwalamukhi, H.P., India
| | - Pravin Kumar
- Department of Pharmaceutics, Laureate Institute of Pharmacy, Kathog, Jwalamukhi, H.P., India
| |
Collapse
|
3
|
Das S, Ghosh B, Sahoo RN, Nayak AK. Recent Advancements in Bioelectronic Medicine: A Review. Curr Drug Deliv 2024; 21:1445-1459. [PMID: 38173212 DOI: 10.2174/0115672018286832231218112557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
Bioelectronic medicine is a multidisciplinary field that combines molecular medicine, neurology, engineering, and computer science to design devices for diagnosing and treating diseases. The advancements in bioelectronic medicine can improve the precision and personalization of illness treatment. Bioelectronic medicine can produce, suppress, and measure electrical activity in excitable tissue. Bioelectronic devices modify specific neural circuits using electrons rather than pharmaceuticals and uses of bioelectronic processes to regulate the biological processes underlining various diseases. This promotes the potential to address the underlying causes of illnesses, reduce adverse effects, and lower costs compared to conventional medication. The current review presents different important aspects of bioelectronic medicines with recent advancements. The area of bioelectronic medicine has a lot of potential for treating diseases, enabling non-invasive therapeutic intervention by regulating brain impulses. Bioelectronic medicine uses electricity to control biological processes, treat illnesses, or regain lost capability. These new classes of medicines are designed by the technological developments in the detection and regulation of electrical signaling methods in the nervous system. Peripheral nervous system regulates a wide range of processes in chronic diseases; it involves implanting small devices onto specific peripheral nerves, which read and regulate the brain signaling patterns to achieve therapeutic effects specific to the signal capacity of a particular organ. The potential for bioelectronic medicine field is vast, as it investigates for treatment of various diseases, including rheumatoid arthritis, diabetes, hypertension, paralysis, chronic illnesses, blindness, etc.
Collapse
Affiliation(s)
- Sudipta Das
- Department of Pharmaceutics, Netaji Subhas Chandra Bose Institute of Pharmacy, Chakdaha, Nadia - 741222, West Bengal, India
| | - Baishali Ghosh
- Department of Pharmaceutics, Netaji Subhas Chandra Bose Institute of Pharmacy, Chakdaha, Nadia - 741222, West Bengal, India
| | - Rudra Narayan Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| | - Amit Kumar Nayak
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| |
Collapse
|
4
|
Seifert JA, Bemis EA, Ramsden K, Lowell C, Polinski K, Feser M, Fleischer C, Demoruelle MK, Buckner J, Gregersen PK, Keating RM, Mikuls TR, O’Dell JR, Weisman MH, Deane KD, Norris JM, Steere AC, Holers VM. Association of Antibodies to Prevotella copri in Anti-Cyclic Citrullinated Peptide-Positive Individuals At Risk of Developing Rheumatoid Arthritis and in Patients With Early or Established Rheumatoid Arthritis. Arthritis Rheumatol 2023; 75:507-516. [PMID: 36259174 PMCID: PMC10065886 DOI: 10.1002/art.42370] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/20/2022] [Accepted: 09/13/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Prevotella copri (P copri), a gut commensal, has been reported to be an immune-relevant organism in individuals with rheumatoid arthritis (RA). This study sought to evaluate anti-P copri (anti-Pc) antibody responses in our participant cohorts and to determine when in the natural history of RA such responses develop. METHODS We analyzed serum levels of immunoglobulin A (IgA) and IgG antibodies from a 27-kd protein of P copri (anti-Pc-p27), an immunogenic P copri protein, in study participants at risk of developing RA, participants who transitioned to RA, participants with early RA (<1 year of disease), and participants with established RA, with comparisons made to their matched controls. We also evaluated anti-Pc-p27 antibody levels in individuals stratified by RA-related autoantibody status. RESULTS Overall, participants with RA had significantly higher IgA anti-Pc-p27 antibody levels and trended toward higher IgG anti-Pc-p27 antibody levels compared with matched controls. When stratified by early versus established RA, participants with early RA had median IgG anti-Pc-p27 antibody levels that were overall higher, whereas median IgA anti-Pc-p27 antibody levels were statistically significantly higher in participants with established RA compared with their matched controls. In the autoantibody-specific analyses, the at-risk population with anti-cyclic citrullinated peptide (anti-CCP) antibodies, but not rheumatoid factor (RF), trended toward increased levels of IgG anti-Pc-p27. Additionally, RA participants who were seropositive for both CCP and RF had significantly increased levels of IgA anti-Pc-p27 antibodies and trended toward higher levels of IgG anti-Pc-p27 antibodies compared with matched controls. CONCLUSION Our findings support a potential etiologic role for P copri in both RA preclinical evolution and the subsequent pathogenesis of synovitis.
Collapse
Affiliation(s)
| | | | - Kristina Ramsden
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Cassidy Lowell
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Marie Feser
- University of Colorado Denver, Aurora, Colorado
| | | | | | - Jane Buckner
- Benaroya Research Institute at Virginia Mason, Seattle, Washington
| | - Peter K. Gregersen
- Feinstein Institutes for Medical Research and North Shore-Long Island Jewish Health System, Manhasset, New York
| | | | - Ted R. Mikuls
- University of Nebraska Medical Center and VA Nebraska-Western Iowa Healthcare System, Omaha, Nebraska
| | - James R. O’Dell
- University of Nebraska Medical Center and VA Nebraska-Western Iowa Healthcare System, Omaha, Nebraska
| | | | | | | | - Allen C. Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
5
|
de Jong TA, Semmelink JF, Denis SW, van de Sande MGH, Houtkooper RHL, van Baarsen LGM. Altered lipid metabolism in synovial fibroblasts of individuals at risk of developing rheumatoid arthritis. J Autoimmun 2023; 134:102974. [PMID: 36512907 DOI: 10.1016/j.jaut.2022.102974] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) can augment the inflammatory process observed in synovium of patients with rheumatoid arthritis (RA). A recent transcriptomic study in synovial biopsies revealed changes in metabolic pathways before disease onset in absence of synovial tissue inflammation. This raises the question whether alterations in cellular metabolism in tissue resident FLS underlie disease pathogenesis. MATERIALS AND METHODS To study this, we compared the metabolic profile of FLS isolated from synovial biopsies from individuals with arthralgia who were autoantibody positive but without any evidence of arthritis (RA-risk individuals, n = 6) with FLS from patients with RA (n = 6), osteoarthritis (OA, n = 6) and seronegative controls (n = 6). After synovial digestion, FLS were cultured in vitro and cellular metabolism was assessed using quantitative PCR, flow cytometry, XFe96 Seahorse Analyzer and tritium-labelled oleate oxidation assays. RESULTS Real-time metabolic profiling revealed that basal (p < 0.0001) and maximum mitochondrial respiration (p = 0.0024) were significantly lower in RA FLS compared with control FLS. In all donors, basal respiration was largely dependent on fatty acid oxidation while glucose was only highly used by FLS from RA patients. Moreover, we showed that RA-risk and RA FLS are less metabolically flexible. Strikingly, mitochondrial fatty acid β-oxidation was significantly impaired in RA-risk (p = 0.001) and RA FLS (p < 0.0001) compared with control FLS. CONCLUSION Overall, this study showed several metabolic alterations in FLS even in absence of synovial inflammation, suggesting that these alterations already start before clinical manifestation of disease and may drive disease pathogenesis.
Collapse
Affiliation(s)
- T A de Jong
- Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands; Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, the Netherlands
| | - J F Semmelink
- Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands; Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, the Netherlands
| | - S W Denis
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
| | - M G H van de Sande
- Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands; Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, the Netherlands
| | - R H L Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - L G M van Baarsen
- Department of Rheumatology & Clinical Immunology and Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands; Amsterdam Rheumatology & Immunology Center (ARC), Academic Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Synovial gene signatures associated with the development of rheumatoid arthritis in at risk individuals: A prospective study. J Autoimmun 2022; 133:102923. [PMID: 36208493 DOI: 10.1016/j.jaut.2022.102923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 12/07/2022]
Abstract
OBJECTIVE To identify molecular changes in synovium before arthritis development in individuals at risk of developing rheumatoid arthritis (RA). MATERIALS AND METHODS We included 67 IgM rheumatoid factor and/or anti-citrullinated protein antibody positive individuals with arthralgia but without arthritis. Synovial biopsies were collected after which individuals were prospectively followed for at least 2 years during which 17 developed arthritis. An exploratory genome-wide transcriptional profiling study was performed in 13 preselected individuals to identify transcripts associated with arthritis development (n = 6). Findings were validated using quantitative real-time PCR and immunohistochemistry in the total cohort. RESULTS Microarray-based survival analyses identified 5588 transcripts whose expression levels in synovium were significantly associated with arthritis development. Pathway analysis revealed that synovial tissue of at risk individuals who later developed arthritis display higher expression of genes involved in adaptive immune response-related pathways compared to at risk individuals who did not develop arthritis. Lower expression was observed for genes involved in extracellular matrix receptor interaction, Wnt-mediated signal transduction and lipid metabolism. Two-way hierarchical clustering analyses of a 27-gene signature separated the total at risk cohort into two groups, where pre-RA individuals preferred to cluster together. Immunohistochemistry studies revealed more podoplanin positive cells and lower lipid droplet staining in synovial tissue from pre-RA individuals. CONCLUSION Synovial alterations in adaptive immune response and lipid metabolism are associated with future development of arthritis. Since this data show synovial changes without overt cellular infiltration, these may be attributed to preclinical changes in resident synovial tissue cells such as fibroblasts, macrophages and tissue resident T cells.
Collapse
|
7
|
Wang DW, Pang XT, Zhang H, Gao HX, Leng YF, Chen FQ, Zhang R, Feng Y, Sun ZL. Gut microbial dysbiosis in rheumatoid arthritis: a systematic review protocol of case-control studies. BMJ Open 2022; 12:e052021. [PMID: 35365513 PMCID: PMC8977794 DOI: 10.1136/bmjopen-2021-052021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 02/26/2022] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) has a huge societal impact due to the high prevalence, irreversible joint damage and systemic complications. Gut microbiota plays an important role in the pathogenesis and progression of RA by regulating the host immune system. Restoring intestinal homeostasis by altering the microbiota could be an attractive strategy for the prevention and treatment of RA. However, the signature features of microbial dysbiosis in RA are still controversial. Therefore, we aim to elucidate the characteristic change in the diversity and composition of gut microbiota in RA. METHODS AND ANALYSIS We will systematically search through PubMed, EMBASE, Web of Science and Cochrane Library, as well as dissertations and conference proceedings. The reference lists of all included studies will be also reviewed to retrieve additional relevant studies. The case-control studies that reported either the relative abundance of bacteria at the phylum or genus level or at least one of the alpha-diversity, beta-diversity indexes in both RA and healthy controls will be included. Eligible studies will be screened independently by two reviewers according to the inclusion criteria. The Newcastle-Ottawa Quality Assessment Scale will be used to assess the quality of the included studies. Data extraction, qualitative and quantitative analysis will be performed within the gut microbial dysbiosis in RA. The expected outcomes will be the identification of the specific changes in composition and diversity of the gut microbiota in patients with RA. The quality of evidence will be assessed by the Grading of Recommendations Assessment, Development and Evaluation framework. ETHICS AND DISSEMINATION Ethical approval is unnecessary as this review does not address the data and privacy of patients. The results will be published in a peer-reviewed scientific journal and conference presentations. PROSPERO REGISTRATION NUMBER CRD42021225229.
Collapse
Affiliation(s)
- Dan-Wen Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xiang-Tian Pang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Heng Zhang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Hai-Xia Gao
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yu-Fei Leng
- Animal Surgery Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Qin Chen
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Rui Zhang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yun Feng
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zhi-Ling Sun
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
8
|
Eriksson K, Lundmark A, Delgado LF, Hu YOO, Fei G, Lee L, Fei C, Catrina AI, Jansson L, Andersson AF, Yucel-Lindberg T. Salivary Microbiota and Host-Inflammatory Responses in Periodontitis Affected Individuals With and Without Rheumatoid Arthritis. Front Cell Infect Microbiol 2022; 12:841139. [PMID: 35360114 PMCID: PMC8964114 DOI: 10.3389/fcimb.2022.841139] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives Periodontitis and rheumatoid arthritis (RA) are two widespread chronic inflammatory diseases with a previously suggested association. The objective of the current study was to compare the oral microbial composition and host´s inflammatory mediator profile of saliva samples obtained from subjects with periodontitis, with and without RA, as well as to predict biomarkers, of bacterial pathogens and/or inflammatory mediators, for classification of samples associated with periodontitis and RA. Methods Salivary samples were obtained from 53 patients with periodontitis and RA and 48 non-RA with chronic periodontitis. The microbial composition was identified using 16S rRNA gene sequencing and compared across periodontitis patients with and without RA. Levels of inflammatory mediators were determined using a multiplex bead assay, compared between the groups and correlated to the microbial profile. The achieved data was analysed using PCoA, DESeq2 and two machine learning algorithms, OPLS-DA and sPLS-DA. Results Differential abundance DESeq2 analyses showed that the four most highly enriched (log2 FC >20) amplicon sequence variants (ASVs) in the non-RA periodontitis group included Alloprevotella sp., Prevotella sp., Haemophilus sp., and Actinomyces sp. whereas Granulicatella sp., Veillonella sp., Megasphaera sp., and Fusobacterium nucleatum were the most highly enriched ASVs (log2 FC >20) in the RA group. OPLS-DA with log2 FC analyses demonstrated that the top ASVs with the highest importance included Vampirovibrio sp. having a positive correlation with non-RA group, and seven ASVs belonging to Sphingomonas insulae, Sphingobium sp., Novosphingobium aromaticivorans, Delftia acidovorans, Aquabacterium spp. and Sphingomonas echinoides with a positive correlation with RA group. Among the detected inflammatory mediators in saliva samples, TWEAK/TNFSF12, IL-35, IFN-α2, pentraxin-3, gp130/sIL6Rb, sIL-6Ra, IL-19 and sTNF-R1 were found to be significantly increased in patients with periodontitis and RA compared to non-RA group with periodontitis. Moreover, correlations between ASVs and inflammatory mediators using sPLS-DA analysis revealed that TWEAK/TNFSF12, pentraxin-3 and IL-19 were positively correlated with the ASVs Sphingobium sp., Acidovorax delafieldii, Novosphingobium sp., and Aquabacterium sp. Conclusion Our results suggest that the combination of microbes and host inflammatory mediators could be more efficient to be used as a predictable biomarker associated with periodontitis and RA, as compared to microbes and inflammatory mediators alone.
Collapse
Affiliation(s)
- Kaja Eriksson
- Department of Dental Medicine, Division of Pediatric Dentistry, Karolinska Institutet, Huddinge, Sweden
- *Correspondence: Kaja Eriksson, ; Tülay Yucel-Lindberg,
| | - Anna Lundmark
- Department of Dental Medicine, Division of Pediatric Dentistry, Karolinska Institutet, Huddinge, Sweden
| | - Luis F. Delgado
- KTH Royal Institute of Technology, Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
| | - Yue O. O. Hu
- Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research, Karolinska Institutet, Stockholm, Sweden
| | - Guozhong Fei
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Region, Stockholm, Sweden
| | - Linkiat Lee
- Department of Dental Medicine, Division of Pediatric Dentistry, Karolinska Institutet, Huddinge, Sweden
| | - Carina Fei
- Department of Dental Medicine, Division of Pediatric Dentistry, Karolinska Institutet, Huddinge, Sweden
| | - Anca I. Catrina
- Rheumatology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Leif Jansson
- Department of Dental Medicine, Division of Periodontology, Karolinska Institutet, Huddinge, Sweden
- Department of Periodontology, Folktandvården Stockholms län AB, Folktandvården Eastmaninstitutet, Stockholm, Sweden
| | - Anders F. Andersson
- KTH Royal Institute of Technology, Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Stockholm, Sweden
| | - Tülay Yucel-Lindberg
- Department of Dental Medicine, Division of Pediatric Dentistry, Karolinska Institutet, Huddinge, Sweden
- *Correspondence: Kaja Eriksson, ; Tülay Yucel-Lindberg,
| |
Collapse
|
9
|
Falahee M, Simons G, DiSantostefano RL, Valor Méndez L, Radawski C, Englbrecht M, Schölin Bywall K, Tcherny-Lessenot S, Kihlbom U, Hauber B, Veldwijk J, Raza K. Treatment preferences for preventive interventions for rheumatoid arthritis: protocol of a mixed methods case study for the Innovative Medicines Initiative PREFER project. BMJ Open 2021; 11:e045851. [PMID: 36916312 PMCID: PMC8039213 DOI: 10.1136/bmjopen-2020-045851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/09/2021] [Accepted: 03/24/2021] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Amidst growing consensus that stakeholder decision-making during drug development should be informed by an understanding of patient preferences, the Innovative Medicines Initiative project 'Patient Preferences in Benefit-Risk Assessments during the Drug Life Cycle' (PREFER) is developing evidence-based recommendations about how and when patient preferences should be integrated into the drug life cycle. This protocol describes a PREFER clinical case study which compares two preference elicitation methodologies across several populations and provides information about benefit-risk trade-offs by those at risk of rheumatoid arthritis (RA) for preventive interventions. METHODS AND ANALYSIS This mixed methods study will be conducted in three countries (UK, Germany, Romania) to assess preferences of (1) first-degree relatives (FDRs) of patients with RA and (2) members of the public. Focus groups using nominal group techniques (UK) and ranking surveys (Germany and Romania) will identify and rank key treatment attributes. Focus group transcripts will be analysed thematically using the framework method and average rank orders calculated. These results will inform the treatment attributes to be assessed in a survey including a discrete choice experiment (DCE) and a probabilistic threshold technique (PTT). The survey will also include measures of sociodemographic variables, health literacy, numeracy, illness perceptions and beliefs about medicines. The survey will be administered to (1) 400 FDRs of patients with RA (UK); (2) 100 FDRs of patients with RA (Germany); and (3) 1000 members of the public in each of UK, Germany and Romania. Logit-based approaches will be used to analyse the DCE and imputation and interval regression for the PTT. ETHICS AND DISSEMINATION This study has been approved by the London-Hampstead Research Ethics Committee (19/LO/0407) and the Ethics Committee of the Friedrich-Alexander-Universität Erlangen-Nürnberg (92_17 B). The protocol has been approved by the PREFER expert review board. The results will be disseminated widely and will inform the PREFER recommendations.
Collapse
Affiliation(s)
- Marie Falahee
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Gwenda Simons
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | - Larissa Valor Méndez
- Department of Internal Medicine and Institute for Clinical Immunology, Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | | | | | | | | | - Ulrik Kihlbom
- Centre for Research Ethics & Bioethics, Uppsala University, Uppsala, Sweden
| | - Brett Hauber
- Health Preference Assessment, RTI Health Solutions, Research Triangle Park, North Carolina, USA
- Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, University of Washington School of Pharmacy, Seattle, Washington, USA
| | - Jorien Veldwijk
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- School of Health Policy & Management and Erasmus Choice Modelling Centre, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Karim Raza
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Department of Rheumatology, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research and Research into Inflammatory Arthritis Centre Versus Arthritis, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
10
|
The pre-clinical phase of rheumatoid arthritis: From risk factors to prevention of arthritis. Autoimmun Rev 2021; 20:102797. [PMID: 33746022 DOI: 10.1016/j.autrev.2021.102797] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease considered as a multistep process spanning from the interaction of genetic (e.g., shared epitope or non-HLA loci), environmental and behavioral risk factors (e.g., smoking) leading to breaking immune tolerance and autoimmune processes such as the production of autoantibodies (e.g., antibodies against citrullinated proteins ACPA or rheumatoid factors, RF), development of the first symptoms without clinical arthritis, and, finally, the manifestation of arthritis. Despite the typical joint involvement in established RA, the pathogenesis of the disease likely begins far from joint structures: in the lungs or periodontium in association with citrullination, intestinal microbiome, or adipose tissue, which supports normal findings in synovial tissue in ACPA+ patients with arthralgia. The presence of ACPA is detectable even years before the first manifestation of RA. The pre-clinical phase of RA is the period preceding clinically apparent RA with ACPA contributing to the symptoms without subclinical inflammation. While the combination of ACPA and RF increases the risk of progression to RA by up to 10 times, increasing numbers of novel autoantibodies are to be investigated to contribute to the increased risk and pathogenesis of RA. With growing knowledge about the course of RA, new aspiration emerges to cure and even prevent RA, shifting the "window of opportunity" to the pre-clinical phases of RA. The clinical definition of individuals at risk of developing RA (clinically suspect arthralgia, CSA) makes it possible to unify these at-risk individuals' clinical characteristics for "preventive" treatment in ongoing clinical trials using mostly biological or conventional synthetic disease-modifying drugs. However, the combination of symptoms, laboratory, and imaging biomarkers may be the best approach to select the correct target at-risk population. The current review aims to explore different phases of RA and discuss the potential of (non)pharmacological intervention aiming to prevent RA.
Collapse
|
11
|
van Boheemen L, Ter Wee MM, Seppen B, van Schaardenburg D. How to enhance recruitment of individuals at risk of rheumatoid arthritis into trials aimed at prevention: understanding the barriers and facilitators. RMD Open 2021; 7:e001592. [PMID: 33685929 PMCID: PMC7942248 DOI: 10.1136/rmdopen-2021-001592] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Several trials to test the efficacy of a pharmacological intervention aimed at primary prevention of rheumatoid arthritis (RA) are ongoing or have recently been completed. A common issue in these trials is the severe difficulty with patient recruitment. In order to enhance recruitment, this qualitative study identified barriers and facilitators of individuals at risk of RA to participate in a prevention trial. METHODS Individuals at risk of developing RA (ie, arthralgia with anticitrullinated protein antibodies and/or rheumatoid factor without arthritis), who had previously been asked to participate in a prevention trial, participated in focus group discussions (n=18) exploring their facilitators and barriers for trial participation. Thematic analysis identified factors that were important in at-risk individuals' decision about trial participation. RESULTS The prospect of personal benefit, the acknowledgement of one's symptoms and the desire to contribute to society facilitated trial participation. In contrast, misconception about what it means to be at risk, or about the aim of the prevention trial, negative views on trial medication, and a low perceived urgency to act on the possibility of developing RA versus a high perceived burden of participating in a trial discouraged participation. CONCLUSIONS To enhance inclusion in trials aimed to prevent RA, the results suggest to use strategies such as optimising education about RA, personal risk, trial aim and trial medication, explicitly addressing misconceptions and concerns, using tools to improve information provision, limiting study burden in trial design and encouraging physicians to mention trial participation.
Collapse
Affiliation(s)
- Laurette van Boheemen
- Rheumatology, Reade, Amsterdam Rheumatology and Immunology Center, Amsterdam, North Holland, The Netherlands
| | - Marieke M Ter Wee
- Epidemiology and Data Science, Amsterdam Public Health, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
- Rheumatology and immunology, AI&I, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Bart Seppen
- Rheumatology, Reade, Amsterdam Rheumatology and Immunology Center, Amsterdam, North Holland, The Netherlands
| | - Dirkjan van Schaardenburg
- Rheumatology, Reade, Amsterdam Rheumatology and Immunology Center, Amsterdam, North Holland, The Netherlands
- Rheumatology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Oliveira IM, Gonçalves C, Oliveira EP, Simón-Vázquez R, da Silva Morais A, González-Fernández Á, Reis RL, Oliveira JM. PAMAM dendrimers functionalised with an anti-TNF α antibody and chondroitin sulphate for treatment of rheumatoid arthritis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111845. [PMID: 33579481 DOI: 10.1016/j.msec.2020.111845] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 12/18/2020] [Accepted: 12/28/2020] [Indexed: 01/22/2023]
|
13
|
Rodríguez-Carrio J, Coras R, Alperi-López M, López P, Ulloa C, Ballina-García FJ, Armando AM, Quehenberger O, Guma M, Suárez A. Profiling of Serum Oxylipins During the Earliest Stages of Rheumatoid Arthritis. Arthritis Rheumatol 2020; 73:401-413. [PMID: 33001576 DOI: 10.1002/art.41537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/24/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Eicosanoids modulate inflammation via complex networks involving different pathways and downstream mediators, including oxylipins. Although altered eicosanoids are linked to rheumatoid arthritis (RA), suggesting that metabolization is enhanced, the role of oxylipins in disease stratification remains unexplored. This study was undertaken to characterize oxylipin networks during the earliest stages of RA and evaluate their associations with clinical features and treatment outcomes. METHODS In total, 60 patients with early RA (according to the American College of Rheumatology/European League Against Rheumatism 2010 criteria), 11 individuals with clinically suspect arthralgia (CSA), and 28 healthy control subjects were recruited. Serum samples were collected at the time of onset. In the early RA group, 50 patients who had not been exposed to disease-modifying antirheumatic drug (DMARD) or glucocorticoid treatment at the time of recruitment were prospectively followed up at 6 and 12 months after having received conventional synthetic DMARDs. A total of 75 oxylipins, mostly derived from arachidonic, eicosapentanoic, and linoleic acids, were identified in the serum by liquid chromatography tandem mass spectrometry. RESULTS Univariate analyses demonstrated differences in expression patterns of 14 oxylipins across the RA, CSA, and healthy control groups, with each exhibiting a different trajectory. Network analyses revealed a strong grouping pattern of oxylipins in RA patients, whereas in individuals with CSA, a fuzzy network of oxylipins with higher degree and closeness was found. Partial least-squares discriminant analyses yielded variable important projection scores of >1 for 22 oxylipins, which allowed the identification of 2 clusters. Cluster usage differed among the groups (P = 0.003), and showed associations with disease severity and low rates of remission at 6 and 12 months in RA patients who were initially treatment-naive. Pathway enrichment analyses revealed different precursors and pathways between the groups, highlighting the relevance of the arachidonic acid pathway in individuals with CSA and the lipooxygenase pathway in patients with early RA. In applying distinct oxylipin signatures, subsets of seropositive and seronegative RA could be identified. CONCLUSION Oxylipin networks differ across stages during the earliest phases of RA. These distinct oxylipin networks could potentially elucidate pathways with clinical relevance for disease progression, clinical heterogeneity, and treatment response.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Universidad de Oviedo, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, and Instituto Reina Sofía de Investigación Nefrológica, REDinREN del Instituto de Salud Carlos III, Oviedo, Spain
| | - Roxana Coras
- University of California School of Medicine, San Diego, and Autonomous University of Barcelona, Barcelona, Spain
| | - Mercedes Alperi-López
- Instituto de Investigación Sanitaria del Principado de Asturias and Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patricia López
- Instituto de Investigación Sanitaria del Principado de Asturias and Universidad de Oviedo, Oviedo, Spain
| | - Catalina Ulloa
- Instituto de Investigación Sanitaria del Principado de Asturias, Instituto Reina Sofía de Investigación Nefrológica, REDinREN del Instituto de Salud Carlos III, and Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Francisco Javier Ballina-García
- Instituto de Investigación Sanitaria del Principado de Asturias and Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | - Mónica Guma
- University of California School of Medicine, San Diego, and Autonomous University of Barcelona, Barcelona, Spain
| | - Ana Suárez
- Instituto de Investigación Sanitaria del Principado de Asturias and Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
14
|
van Boheemen L, Bolt JW, Ter Wee MM, de Jong HM, van de Sande MG, van Schaardenburg D. Patients' and rheumatologists' perceptions on preventive intervention in rheumatoid arthritis and axial spondyloarthritis. Arthritis Res Ther 2020; 22:217. [PMID: 32933547 PMCID: PMC7493385 DOI: 10.1186/s13075-020-02314-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/07/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Individuals at risk of developing rheumatoid arthritis (RA) may benefit from lifestyle or pharmacological interventions aimed at primary prevention. The same may apply to individuals at risk of axial spondyloarthritis (axSpA). Our aim was to investigate and compare the willingness of individuals at risk of RA or axSpA and rheumatologists to initiate preventive intervention. METHODS Individuals at risk of RA (arthralgia and anti-citrullinated protein antibodies and/or rheumatoid factor positivity without arthritis (RA-risk cohort; n = 100)), axSpA (first-degree relatives of HLA-B27-positive axSpA patients (SpA-risk cohort; n = 38)), and Dutch rheumatologists (n = 49) completed a survey on preventive intervention which included questions about disease perception, lifestyle intervention, and preventive medication. RESULTS At-risk individuals reported willingness to change median 7 of 13 lifestyle components in the areas of smoking, diet, and exercise. In contrast, 35% of rheumatologists gave lifestyle advice to ≥ 50% of at-risk patients. The willingness to use 100% effective preventive medication without side effects was 53% (RA-risk), 55% (SpA-risk), and 74% (rheumatologists) at 30% disease risk which increased to 69% (RA-risk) and 92% (SpA-risk and rheumatologists) at 70% risk. With minor side effects, willingness was 26%, 29%, and 31% (at 30% risk) versus 40%, 66%, and 76% (at 70% risk), respectively. CONCLUSIONS Risk perception and willingness to start preventive intervention were largely similar between individuals at risk of RA and axSpA. Although the willingness to change lifestyle is high among at-risk individuals, most rheumatologists do not advise them to change their lifestyle. In contrast, rheumatologists are more willing than at-risk patients to start preventive medication.
Collapse
Affiliation(s)
- Laurette van Boheemen
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, PO box 58271, 1040 HG, Amsterdam, the Netherlands.
| | - Janne W Bolt
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marieke M Ter Wee
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, VUmc, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam UMC, VUmc, Amsterdam, the Netherlands
| | - Henriëtte M de Jong
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marleen G van de Sande
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dirkjan van Schaardenburg
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, PO box 58271, 1040 HG, Amsterdam, the Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
15
|
Mosor E, Stoffer-Marx M, Steiner G, Raza K, Stack RJ, Simons G, Falahee M, Skingle D, Dobrin M, Schett G, Englbrecht M, Smolen JS, Kjeken I, Hueber AJ, Stamm TA. I Would Never Take Preventive Medication! Perspectives and Information Needs of People Who Underwent Predictive Tests for Rheumatoid Arthritis. Arthritis Care Res (Hoboken) 2020; 72:360-368. [PMID: 30710453 PMCID: PMC7064954 DOI: 10.1002/acr.23841] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Little is known about the experiences, values, and needs of people without arthritis who undergo predictive biomarker testing for the development of rheumatoid arthritis (RA). Our study aimed to explore the perspectives of these individuals and describe their information needs. METHODS A qualitative, multicenter interview study with a thematic analysis was conducted in Austria, Germany and the UK. Individuals were interviewed who underwent predictive biomarker testing for RA and had a positive test result but no diagnosis of any inflammatory joint disease. Participants included patients with arthralgia and asymptomatic individuals. Information and education needs were developed from the qualitative codes and themes using the Arthritis Educational Needs Assessment Tool as a frame of reference. RESULTS Thematic saturation was reached in 34 individuals (76% female, 24 [71%] with arthralgia, and 10 [29%] asymptomatic individuals). Thirty-seven codes were summarized into 4 themes: 1) decision-making around whether to undergo initial predictive testing, 2) willingness to consider further predictive tests, and/or 3) preventive interventions, including medication, and 4) varying reactions after receiving a positive test result. Individuals with arthralgia were more likely to be willing to take preventive action, undergo further testing, and experience psychological distress than asymptomatic individuals. All participants expressed the need for tailored, patient-understandable information. CONCLUSION Individuals at risk of RA are currently the subjects of research aimed at developing better predictive strategies and preventive approaches. Their perceptions and needs should be addressed to inform the future development of interventions combined with education.
Collapse
Affiliation(s)
- Erika Mosor
- Medical University of Vienna, Vienna, Austria
| | | | - Günter Steiner
- Medical University of Vienna and Ludwig Boltzmann Cluster Arthritis and Rehabilitation, Vienna, Austria
| | - Karim Raza
- University of Birmingham and Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | | | | | | | | | - Mircia Dobrin
- The Romanian League Against Rheumatism, Timisoara, Romania
| | - Georg Schett
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Matthias Englbrecht
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Ingvild Kjeken
- Oslo Metropolitan University and Diakonhjemmet Hospital, Oslo, Norway
| | - Axel J Hueber
- Friedrich-Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tanja A Stamm
- Medical University of Vienna and Ludwig Boltzmann Cluster Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|
16
|
Greenblatt HK, Kim HA, Bettner LF, Deane KD. Preclinical rheumatoid arthritis and rheumatoid arthritis prevention. Curr Opin Rheumatol 2020; 32:289-296. [PMID: 32205569 PMCID: PMC7340337 DOI: 10.1097/bor.0000000000000708] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW This review is to provide an update on the current understanding of rheumatoid arthritis (RA) development related to disease development prior to the onset clinically apparent synovitis (i.e. Pre-RA), and opportunities for disease prevention. RECENT FINDINGS A growing number of studies have demonstrated that serum elevations of autoantibodies rheumatoid factor, antibodies to citrullinated protein/peptide antigens (ACPAs) and antibodies to other posttranslationally modified proteins (e.g. carbamylated proteins) are highly predictive of future development of inflammatory arthritis/RA during a period that can be termed Pre-RA. Other factors including genetic, environmental, symptoms and imaging findings can also enhance prediction. Moreover, several novel biomarkers and changes in autoantibodies (e.g. glycosylation of variable domains) have been identified in Pre-RA. There has also been growing evidence that initiation and propagation of RA-related autoimmunity during the Pre-RA phase may be related to mucosal processes. The discovery of Pre-RA has also underpinned the development of several clinical prevention trials in RA; specifically, the PRAIRI study demonstrated that a single dose of rituximab can delay the onset of clinically apparent IA in at-risk individuals. Additional studies are evaluating the ability of drugs including abatacept, hydroxychloroquine and methotrexate to prevent or delay future RA. SUMMARY The results from ongoing natural history and prevention trials in RA should further inform several critical issues in RA prevention including identification and enrolment of individuals at high-risk of imminent RA, the efficacy, safety and cost-effectiveness of prevention, and potentially the identification of new targets for prevention.
Collapse
Affiliation(s)
| | - Hyoun-Ah Kim
- University of Colorado Anschutz Medical Campus
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Republic of Korea
| | | | | |
Collapse
|
17
|
Bao M, Zhang K, Wei Y, Hua W, Gao Y, Li X, Ye L. Therapeutic potentials and modulatory mechanisms of fatty acids in bone. Cell Prolif 2020; 53:e12735. [PMID: 31797479 PMCID: PMC7046483 DOI: 10.1111/cpr.12735] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Bone metabolism is a lifelong process that includes bone formation and resorption. Osteoblasts and osteoclasts are the predominant cell types associated with bone metabolism, which is facilitated by other cells such as bone marrow mesenchymal stem cells (BMMSCs), osteocytes and chondrocytes. As an important component in our daily diet, fatty acids are mainly categorized as long-chain fatty acids including polyunsaturated fatty acids (LCPUFAs), monounsaturated fatty acids (LCMUFAs), saturated fatty acids (LCSFAs), medium-/short-chain fatty acids (MCFAs/SCFAs) as well as their metabolites. Fatty acids are closely associated with bone metabolism and associated bone disorders. In this review, we summarized the important roles and potential therapeutic implications of fatty acids in multiple bone disorders, reviewed the diverse range of critical effects displayed by fatty acids on bone metabolism, and elucidated their modulatory roles and mechanisms on specific bone cell types. The evidence supporting close implications of fatty acids in bone metabolism and disorders suggests fatty acids as potential therapeutic and nutritional agents for the treatment and prevention of metabolic bone diseases.
Collapse
Affiliation(s)
- Minyue Bao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Kaiwen Zhang
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yangyini Wei
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Weihan Hua
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yanzi Gao
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin Li
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesDepartment of Cariology and EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
18
|
Abstract
Multiple studies have shown that there is a pre-clinical period preceding the development of rheumatoid arthritis (RA). During this period, complex interactions between the environmental and genetic causes occur, and the expression "preclinical RA" has been proposed to define it. Early treatment intervention is associated with less joint damage and has an increased possibility of achieving remission. In this review, we provide an overview of the preclinical phases of RA, new immunological and imaging biomarkers, and the clinical features, and the management of individuals at-risk of developing RA.
Collapse
Affiliation(s)
- Patrícia Martins
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Portugal; Unidade de Investigação Em Reumatologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Portugal.
| | - João Eurico Fonseca
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Portugal; Unidade de Investigação Em Reumatologia, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Portugal
| |
Collapse
|
19
|
Raza K, Holers VM, Gerlag D. Nomenclature for the Phases of the Development of Rheumatoid Arthritis. Clin Ther 2019; 41:1279-1285. [PMID: 31196657 DOI: 10.1016/j.clinthera.2019.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/27/2019] [Accepted: 04/10/2019] [Indexed: 12/26/2022]
Abstract
Rheumatoid arthritis (RA) is a common immune-mediated inflammatory disease. Research on RA is increasingly focused on the earliest stages of the disease, and has provided strong evidence that clinical signs and symptoms may be preceded by a preclinical phase during which evidence of systemic autoimmunity may be present. To facilitate research in this area, a number of international initiatives have proposed definitions of the phases of disease leading up to RA. The first of these initiatives was the European League Against Rheumatism's (EULAR) set of recommendations on terminology in persons at risk for RA, which suggested that the "at-risk phases" be described in terms of patients variably having: (A) genetic risk factors for RA; (B) environmental risk factors for RA; (C) systemic autoimmunity associated with RA; (D) symptoms without clinical arthritis; and (E) unclassified arthritis. The phrase clinically suspect arthralgia (CSA) is now widely used and can be regarded as describing a subgroup of patients in phase D. A definition of CSA was recently proposed by a EULAR taskforce, and primary research has begun to explore the full range of symptoms, as well as their sensitivity and specificity alone and in combination with other factors, that characterize this phase. Similarly, immune abnormalities at mucosal and others sites that precede and/or are associated with the onset of musculoskeletal symptoms are being increasingly studied and understood. Whether some of these at-risk phases, in particular CSA, represent entities meriting their own classification criteria is an essential area for consensus and will be discussed.
Collapse
Affiliation(s)
- Karim Raza
- Institute of Inflammation and Ageing, Arthritis Research UK Rheumatoid Arthritis Centre of Excellence, Medical Research Council Arthritis Research UK Centre for Musculoskeletal Ageing, University of Birmingham, Birmingham, United Kingdom; Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom.
| | - V Michael Holers
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | | |
Collapse
|
20
|
|
21
|
Croft AP, Campos J, Jansen K, Turner JD, Marshall J, Attar M, Savary L, Wehmeyer C, Naylor AJ, Kemble S, Begum J, Dürholz K, Perlman H, Barone F, McGettrick HM, Fearon DT, Wei K, Raychaudhuri S, Korsunsky I, Brenner MB, Coles M, Sansom SN, Filer A, Buckley CD. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 2019; 570:246-251. [PMID: 31142839 PMCID: PMC6690841 DOI: 10.1038/s41586-019-1263-7] [Citation(s) in RCA: 617] [Impact Index Per Article: 102.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 05/02/2019] [Indexed: 01/18/2023]
Abstract
The identification of lymphocyte subsets with non-overlapping effector functions has been pivotal to the development of targeted therapies in immune-mediated inflammatory diseases (IMIDs)1,2. However, it remains unclear whether fibroblast subclasses with non-overlapping functions also exist and are responsible for the wide variety of tissue-driven processes observed in IMIDs, such as inflammation and damage3-5. Here we identify and describe the biology of distinct subsets of fibroblasts responsible for mediating either inflammation or tissue damage in arthritis. We show that deletion of fibroblast activation protein-α (FAPα)+ fibroblasts suppressed both inflammation and bone erosions in mouse models of resolving and persistent arthritis. Single-cell transcriptional analysis identified two distinct fibroblast subsets within the FAPα+ population: FAPα+THY1+ immune effector fibroblasts located in the synovial sub-lining, and FAPα+THY1- destructive fibroblasts restricted to the synovial lining layer. When adoptively transferred into the joint, FAPα+THY1- fibroblasts selectively mediate bone and cartilage damage with little effect on inflammation, whereas transfer of FAPα+ THY1+ fibroblasts resulted in a more severe and persistent inflammatory arthritis, with minimal effect on bone and cartilage. Our findings describing anatomically discrete, functionally distinct fibroblast subsets with non-overlapping functions have important implications for cell-based therapies aimed at modulating inflammation and tissue damage.
Collapse
Affiliation(s)
- Adam P Croft
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- Versus Arthritis Centre of Excellence in the Pathogenesis of Rheumatoid Arthritis, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Joana Campos
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Kathrin Jansen
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Jason D Turner
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Jennifer Marshall
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Moustafa Attar
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Loriane Savary
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Corinna Wehmeyer
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- Musculoskeletal Medicine, University of Muenster, Muenster, Germany
| | - Amy J Naylor
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Samuel Kemble
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Jenefa Begum
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Kerstin Dürholz
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Harris Perlman
- Department of Medicine, Division of Rheumatology, Northwestern University, Feinberg School of Medicine Chicago, Evanston, IL, USA
| | - Francesca Barone
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Helen M McGettrick
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | | | - Kevin Wei
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ilya Korsunsky
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark Coles
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Stephen N Sansom
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Filer
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- Versus Arthritis Centre of Excellence in the Pathogenesis of Rheumatoid Arthritis, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- MRC and Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Christopher D Buckley
- Rheumatology Research Group, Institute for Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK.
- Versus Arthritis Centre of Excellence in the Pathogenesis of Rheumatoid Arthritis, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK.
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- MRC and Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK.
| |
Collapse
|
22
|
Hagert C, Siitonen R, Li XG, Liljenbäck H, Roivainen A, Holmdahl R. Rapid spread of mannan to the immune system, skin and joints within 6 hours after local exposure. Clin Exp Immunol 2019; 196:383-391. [PMID: 30712330 DOI: 10.1111/cei.13268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2019] [Indexed: 12/01/2022] Open
Abstract
Psoriasis (Ps), psoriatic arthritis (PsA) and rheumatoid arthritis (RA) are common diseases dependent on environmental factors that activate the immune system in unknown ways. Mannan is a group of polysaccharides common in the environment; they are potentially pathogenic, because at least some of them induce Ps-, PsA- and RA-like inflammation in mice. Here, we used positron emission tomography/computed tomography to examine in-vivo transport and spread of mannan labelled with fluorine-18 [18 F]. The results showed that mannan was transported to joints (knee) and bone marrow (tibia) of mice within 6 h after intraperitoneal injection. The time it took to transport mannan, and its presence in blood, indicated cellular transport of mannan within the circulatory system. In addition, mannan was filtered mainly through the spleen and liver. [18 F]fluoromannan was excreted via kidneys, small intestine and, to some extent, the mouth. In conclusion, mannan reaches joints rapidly after injection, which may explain why mannan-induced inflammatory disease is targeted to these tissues.
Collapse
Affiliation(s)
- C Hagert
- Medical Inflammation Research, MediCity Research Laboratory, University of Turku, Turku, Finland.,The National Doctoral Programme in Informational and Structural Biology, Turku, Finland
| | - R Siitonen
- Turku PET Centre, University of Turku, Turku, Finland
| | - X-G Li
- Turku PET Centre, University of Turku, Turku, Finland.,Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - H Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - A Roivainen
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | - R Holmdahl
- Medical Inflammation Research, MediCity Research Laboratory, University of Turku, Turku, Finland.,Medical Inflammation Research, Department of Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
23
|
Gerlag DM, Safy M, Maijer KI, Tang MW, Tas SW, Starmans-Kool MJF, van Tubergen A, Janssen M, de Hair M, Hansson M, de Vries N, Zwinderman AH, Tak PP. Effects of B-cell directed therapy on the preclinical stage of rheumatoid arthritis: the PRAIRI study. Ann Rheum Dis 2019; 78:179-185. [PMID: 30504445 PMCID: PMC6352407 DOI: 10.1136/annrheumdis-2017-212763] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVES We explored the effects of B-cell directed therapy in subjects at risk of developing autoantibodypositive rheumatoid arthritis (RA), who never experienced inflammatory arthritis before, and explored biomarkers predictive of arthritis development. METHODS Individuals positive for both anti-citrullinated peptide antibodies and rheumatoid factor but without arthritis were included in a randomised, double-blind, placebo-controlled study to receive a single infusion of 1000 mg rituximab or placebo. RESULTS Eighty-one individuals received treatment and were followed up for a mean of 29.0 (0-54) months, during which 30/81 (37%) individuals developed arthritis. The observed risk of developing arthritis in the placebo-treated group was 40%, which was decreased by 55% (HR 0.45, 95% CI 0.154 to 1.322) in the rituximab-treated group at 12 months. Rituximab treatment caused a delay in arthritis development of 12 months compared with placebo treatment at the point when 25% of the subjects had developed arthritis (p<0.0001). Erythrocyte sedimentation rate and the presence of anti-citrullinated α-enolase peptide 1 at baseline were significant predictors of arthritis development. CONCLUSIONS A single infusion of 1000 mg rituximab significantly delays the development of arthritis in subjects at risk of developing RA, providing evidence for the pathogenetic role of B cells in the earliest, prearthritis stage of autoantibody positive RA.
Collapse
Affiliation(s)
- Danielle M Gerlag
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Epidemiology and Bioinformatics, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Mary Safy
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Karen I Maijer
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Man Wai Tang
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Sander W Tas
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | | | - Astrid van Tubergen
- Division of Rheumatology, Department of Medicine, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Matthijs Janssen
- Department of Rheumatology, Rijnstate Hospital, Arnhem, The Netherlands
| | - Maria de Hair
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Monika Hansson
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Niek de Vries
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | | | - Paul P Tak
- Department of Clinical immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Eriksson K, Nise L, Alfredsson L, Catrina AI, Askling J, Lundberg K, Klareskog L, Yucel-Lindberg T. Seropositivity combined with smoking is associated with increased prevalence of periodontitis in patients with rheumatoid arthritis. Ann Rheum Dis 2018; 77:1236-1238. [PMID: 28986364 PMCID: PMC6059048 DOI: 10.1136/annrheumdis-2017-212091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 11/04/2022]
Affiliation(s)
- Kaja Eriksson
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Nise
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Alfredsson
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre of Environmental and Occupational Medicine, Stockholm County Council, Stockholm, Sweden
| | - Anca Irinel Catrina
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Askling
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
- Clinical Epidemiology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Tülay Yucel-Lindberg
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW This review is to provide an update on the current understanding of rheumatoid arthritis (RA) development related to disease development prior to the onset clinically apparent synovitis and opportunities for disease prevention. RECENT FINDINGS A growing number of studies have demonstrated that serum elevations of autoantibodies rheumatoid factor and antibodies to citrullinated protein/peptide antigens (ACPA) are highly predictive of future development of IA/RA. This has underpinned the development of several prevention trials in RA. The full results from most of these prevention trials are pending, but ultimately, they should further inform several critical issues in RA prevention including identification and enrollment of individuals at high risk of imminent RA, the efficacy, safety and cost-effectiveness of prevention, and potentially the identification of new targets for prevention. Results from studies in RA prevention as well as other ongoing natural history studies of RA will help to change the paradigm of how RA is managed, potentially adding prevention to the possibilities for management.
Collapse
Affiliation(s)
- Kevin D Deane
- Division of Rheumatology, University of Colorado School of Medicine, 1775 Aurora Court, Mail Stop B-115, Aurora, CO, 80045, USA.
| |
Collapse
|
26
|
Erlandsson MC, Turkkila M, Siljehult F, Pullerits R, Eriksson C, Rantapää-Dahlqvist S, Bokarewa MI. Survivin improves the early recognition of rheumatoid arthritis among patients with arthralgia: A population-based study within two university cities of Sweden. Semin Arthritis Rheum 2018; 47:778-785. [PMID: 29174794 DOI: 10.1016/j.semarthrit.2017.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/11/2017] [Accepted: 10/30/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES The aim of this study was to validate the use of survivin for preclinical recognition of rheumatoid arthritis (RA) among patients with unexplained arthralgia. METHODS Serum levels of survivin and the arthritis-specific autoantibodies RF and ACPA were measured in total of 5046 patients with musculoskeletal complains during 12 consecutive months in Gothenburg and in Umeå. Among them, 303 arthralgia patients were identified and prospectively followed. RESULTS After 48 months, 12.2% of the arthralgia patients developed RA. Most of RA cases had high serum survivin, which increased the relative risk for RA (RR = 5.90, p = 3 × 10-7). Combination of survivin with autoantibodies was present in only 4.6% of the arthralgia patients and increased further the risk of RA and shortened time to RA development. Presence of any single autoantibody in the survivin-negative patients was associated with a minor risk for RA and had RA-free survival similar to the reference group. CONCLUSION This study shows that measurement of survivin in serum improves estimation of RA risk and prospectively predicts RA development in patients with arthralgia. Survivin may indicate a phase preceding autoantibody production.
Collapse
Affiliation(s)
- Malin C Erlandsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, University of Gothenburg, Box 480, SE 405 30 Gothenburg, Sweden
| | - Minna Turkkila
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, University of Gothenburg, Box 480, SE 405 30 Gothenburg, Sweden
| | - Filip Siljehult
- Department of Public Health and Clinical Medicine, Rheumatology Unit, University of Umeå, Umeå, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, University of Gothenburg, Box 480, SE 405 30 Gothenburg, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Catharina Eriksson
- Department of Public Health and Clinical Medicine, Rheumatology Unit, University of Umeå, Umeå, Sweden; Laboratory of Clinical Immunology University Hospital of Umeå, Umeå, Sweden
| | | | - Maria I Bokarewa
- Department of Rheumatology and Inflammation Research, Sahlgrenska University Hospital, University of Gothenburg, Box 480, SE 405 30 Gothenburg, Sweden.
| |
Collapse
|
27
|
Lingampalli N, Sokolove J, Lahey LJ, Edison JD, Gilliland WR, Holers VM, Deane KD, Robinson WH. Combination of anti-citrullinated protein antibodies and rheumatoid factor is associated with increased systemic inflammatory mediators and more rapid progression from preclinical to clinical rheumatoid arthritis. Clin Immunol 2018; 195:119-126. [PMID: 29842946 DOI: 10.1016/j.clim.2018.05.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 12/26/2022]
Abstract
The development of rheumatoid factor (RF) and/or anti-citrullinated protein antibodies (ACPAs) can be observed years prior to clinical diagnosis of rheumatoid arthritis (RA). Nevertheless, the interaction between these two autoantibodies and their combined effect on development of RA is unclear. We measured RF, cytokines, and ACPA subtypes in serial pre-clinical serum samples collected from 83 US veterans who all developed RA. Levels of cytokines and ACPAs were compared between the following groups: anti-cyclic citrullinated peptide (anti-CCP)-/RF- (double negative), anti-CCP+/RF-, anti-CCP-/RF+, or anti-CCP+/RF+ (double-positive). The double-positive subgroup had significantly higher levels of 20 inflammatory cytokines and 29 ACPA reactivities, and the shortest interval, 1.3 years, between the preclinical sample timepoint and diagnosis of RA. Thus, the combined presence of ACPAs and RF is associated with a more rapid progression to RA, suggesting that anti-CCP+/RF+ individuals have a more advanced preclinical disease state and that the onset of RA may be imminent.
Collapse
Affiliation(s)
- Nithya Lingampalli
- VA Palo Alto Health Care System, 3801 Miranda Ave, C4-191, Palo Alto, CA 94304, United States; Division of Immunology and Rheumatology, Stanford University, 269 Campus Drive, CCSR 4135, Stanford, CA 94305, United States
| | - Jeremy Sokolove
- VA Palo Alto Health Care System, 3801 Miranda Ave, C4-191, Palo Alto, CA 94304, United States; Division of Immunology and Rheumatology, Stanford University, 269 Campus Drive, CCSR 4135, Stanford, CA 94305, United States
| | - Lauren J Lahey
- VA Palo Alto Health Care System, 3801 Miranda Ave, C4-191, Palo Alto, CA 94304, United States; Division of Immunology and Rheumatology, Stanford University, 269 Campus Drive, CCSR 4135, Stanford, CA 94305, United States
| | - Jess D Edison
- Rheumatology Service, Department of Medicine, Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, United States
| | - William R Gilliland
- Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, United States
| | - V Michael Holers
- Division of Rheumatology, University of Colorado Denver School of Medicine, 1775 Aurora Court, Aurora, CO 80045, United States
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Denver School of Medicine, 1775 Aurora Court, Aurora, CO 80045, United States.
| | - William H Robinson
- VA Palo Alto Health Care System, 3801 Miranda Ave, C4-191, Palo Alto, CA 94304, United States; Division of Immunology and Rheumatology, Stanford University, 269 Campus Drive, CCSR 4135, Stanford, CA 94305, United States.
| |
Collapse
|
28
|
Hagert C, Sareila O, Kelkka T, Jalkanen S, Holmdahl R. The Macrophage Mannose Receptor Regulate Mannan-Induced Psoriasis, Psoriatic Arthritis, and Rheumatoid Arthritis-Like Disease Models. Front Immunol 2018; 9:114. [PMID: 29467756 PMCID: PMC5808283 DOI: 10.3389/fimmu.2018.00114] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/15/2018] [Indexed: 12/24/2022] Open
Abstract
The injection of mannan into mice can result in the development of psoriasis (Ps) and psoriatic arthritis (PsA), whereas co-injection with antibodies toward collagen type II leads to a chronic rheumatoid-like arthritis. The critical event in all these diseases is mannan-mediated activation of macrophages, causing more severe disease if the macrophages are deficient in neutrophil cytosolic factor 1 (Ncf1), i.e., lack the capacity to make a reactive oxygen species (ROS) burst. In this study, we investigated the role of one of the receptors binding mannan; the macrophage mannose receptor (MR, CD206). MR is a C-type lectin present on myeloid cells and lymphatics. We found that mice deficient in MR expression had more severe mannan-induced Ps, PsA as well as rheumatoid-like arthritis. Interestingly, the MR-mediated protection was partly lost in Ncf1 mutated mice and was associated with an type 2 macrophage expansion. In conclusion, these results show that MR protects against a pathogenic inflammatory macrophage response induced by mannan and is associated with induction of ROS.
Collapse
Affiliation(s)
- Cecilia Hagert
- Medicity Research Laboratory, University of Turku, Turku, Finland.,The National Doctoral Programme in Informational and Structural Biology (ISB), Turku, Finland
| | - Outi Sareila
- Medicity Research Laboratory, University of Turku, Turku, Finland.,Medical Inflammation Research, Karolinska Institutet, Stockholm, Sweden
| | - Tiina Kelkka
- Medicity Research Laboratory, University of Turku, Turku, Finland.,The Turku Doctoral Programme of Biomedical Sciences (TuBS), Turku, Finland
| | - Sirpa Jalkanen
- Medicity Research Laboratory, University of Turku, Turku, Finland
| | - Rikard Holmdahl
- Medicity Research Laboratory, University of Turku, Turku, Finland.,Medical Inflammation Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Sakr BR, Elfishawi MM, ElArousy MH, Hatw AK, AbdulKarim AN, Tammam AB, Kotp AN, Hamed ME, Genedy IE, Desouky EDE, Nawito ZO. Rheumatoid arthritis: A single-center Egyptian experience. Immunol Invest 2018; 47:293-302. [DOI: 10.1080/08820139.2018.1425700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- B. R Sakr
- Rheumatology and Rehabilitation Department, Kasr Al-Ainy Hospital, Cairo University , Cairo, Egypt
| | - M. M Elfishawi
- Rheumatology and Rehabilitation Department, Kasr Al-Ainy Hospital, Cairo University , Cairo, Egypt
| | - M. H ElArousy
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - A. K Hatw
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - A. N AbdulKarim
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - A. B Tammam
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - A. N Kotp
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - M. E Hamed
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - I. E Genedy
- Medical students, faculty of medicine, Cairo University , Cairo, Egypt
| | - E. D. El Desouky
- Epidemiology and Biostatistics Department, National Cancer Institute, Cairo University , Cairo, Egypt
| | - Z. O. Nawito
- Rheumatology and Rehabilitation Department, Kasr Al-Ainy Hospital, Cairo University , Cairo, Egypt
| |
Collapse
|
30
|
Poncet’s Disease in the Preclinical Phase of Rheumatoid Arthritis. Case Rep Rheumatol 2018; 2018:3571247. [PMID: 29854537 PMCID: PMC5964554 DOI: 10.1155/2018/3571247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/12/2018] [Accepted: 04/17/2018] [Indexed: 11/17/2022] Open
Abstract
We report on a patient with seropositive polyarthritis retrospectively diagnosed as Poncet’s disease in the preclinical phase of seropositive rheumatoid arthritis. Our patient developed rheumatoid arthritis more than 2 years after being successfully treated for pulmonary tuberculosis and an initial inflammatory polyarthritis consistent with the diagnosis of Poncet’s disease. This case illustrates the importance of recognizing Poncet’s disease in a patient presenting with polyarthritis in order to avoid inappropriate long-term disease modifying antirheumatic treatment. It also illustrates the need for adequate follow-up of patients with Poncet’s disease after treatment with antituberculosis treatment so that progression to a primary inflammatory arthritis such as rheumatoid arthritis may be identified timeously. Although seropositivity for rheumatoid arthritis has been reported in Poncet’s disease as well as in tuberculosis, it is rather uncommon, and long-term follow-up of patients with Poncet’s disease is essential particularly if they have positive serological tests for rheumatoid arthritis. In this case report, we describe the first reported case of Poncet’s disease in the preclinical phase of rheumatoid arthritis and review the literature related to this rare disease presentation.
Collapse
|
31
|
Tak PP, Doorenspleet ME, de Hair MJH, Klarenbeek PL, van Beers-Tas MH, van Kampen AHC, van Schaardenburg D, Gerlag DM, Baas F, de Vries N. Dominant B cell receptor clones in peripheral blood predict onset of arthritis in individuals at risk for rheumatoid arthritis. Ann Rheum Dis 2017; 76:1924-1930. [PMID: 28790026 PMCID: PMC5705849 DOI: 10.1136/annrheumdis-2017-211351] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The onset of seropositive rheumatoid arthritis (RA) is preceded by the presence of specific autoantibodies in the absence of synovial inflammation. Only a subset of these at-risk individuals will develop clinical disease. This impedes efforts to implement early interventions that may prevent onset of clinically manifest disease. Here we analyse whether clonal changes in the B cell receptor (BCR) repertoire can reliably predict onset of signs and symptoms. METHODS In a prospective cohort study in 21 individuals at risk for RA based on the presence of autoantibodies, the BCR repertoire of paired peripheral blood and synovial tissue samples was analysed using next-generation BCR sequencing. BCR clones that were expanded beyond 0.5% of the total repertoire were labelled dominant. The relative risk (RR) for onset of arthritis was assessed using the presence of ≥5 dominant BCR clones as cut-off. Findings in peripheral blood were validated in an independent prospective cohort of 50 at-risk individuals. Based on the test cohort, individuals in the validation cohort were considered positive if peripheral blood at study entry showed ≥5 dominant BCR clones. FINDINGS Both in the test and validation cohort, the presence of ≥5 dominant BCR clones in peripheral blood was significantly associated with arthritis development after follow-up (validation cohort RR 6.3, 95% CI 2.7 to 15, p<1×10-4). Even when adjusted for a recently described clinical prediction rule the association remained intact (RR 5.0, 95% CI 1.2 to 20, p=0.024). When individuals developed arthritis, dominant BCR clones disappeared from peripheral blood and appeared in synovial tissue, suggesting a direct role of these clones in disease pathogenesis. INTERPRETATION Dominant BCR clones in peripheral blood predict onset of clinical signs and symptoms of RA in at-risk individuals with high accuracy. Our data suggest that during onset of RA these clones shift from peripheral blood to the target tissue.
Collapse
Affiliation(s)
- Paul P Tak
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marieke E Doorenspleet
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, Amsterdam, The Netherlands
- Department of Genome Analysis, Academic Medical Center, Amsterdam, The Netherlands
| | - Maria J H de Hair
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul L Klarenbeek
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, Amsterdam, The Netherlands
- Department of Genome Analysis, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Antoine H C van Kampen
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Danielle M Gerlag
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank Baas
- Department of Genome Analysis, Academic Medical Center, Amsterdam, The Netherlands
| | - Niek de Vries
- Amsterdam Rheumatology and Immunology Center, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Abstract
Rheumatoid arthritis (RA) is a chronic immunoinflammatory (autoimmune) disease manifested by progressive joint destruction, systemic inflammation of the internal organs, and a wide range of comorbidities associated with chronic inflammation and frequently with adverse drug reactions. However, despite the major advances in the early diagnosis and treatment of RA, which have led to the radical improvement of prognosis in many patients, the problem of pharmacotherapy for RA is far from being solved. This is determined by a lack of sensitive and specific diagnostic and prognostic biomarkers in the early stage of the disease and, most importantly, by the heterogeneity of immunopathogenesis mechanisms in both at the onset of RA and during its progression, which make the personalization of therapy difficult in the patients. Selective block of inflammatory mediators with innovative medicines is frequently associated with primary inefficiency, secondary drug resistance, the development of generalized immunosuppression, the paradoxical activation of an autoimmune process, and the aggravation of comorbidities. At the same time, it is difficult to search for new RA pharmacotherapy targets since the nature of immunopathological disorders in patients can be substantially different from the inflammatory process that takes place when simulating arthritis in laboratory animals. The paper discusses the novel drugs that are used in rheumatology to treat RA or tested in different phases of preclinical or clinical trials, such as tumor necrosis factor-α inhibitors, interleukin-6 (IL-6), IL-17, anti-B cell therapy, bispecific antibodies, blockers of JAK (and other signaling molecules), bioelectronic vagus nerve activation, dendritic cell-based immunotherapy, and other therapies, as well as approaches to secondary prevention of RA in patients with undifferentiated arthritis and clinically suspect arthralgia, who are at high risk for RA. Decoding the mechanisms underlying the pathogenesis of RA and a chronic inflammatory process as a whole has created preconditions for the design of novel medications for the prevention and treatment of this disease, the introduction of which into clinical practice should lead to a radical improvement of prognosis in this disease.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology
I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| |
Collapse
|
33
|
Alpízar-Rodríguez D, Pluchino N, Canny G, Gabay C, Finckh A. The role of female hormonal factors in the development of rheumatoid arthritis. Rheumatology (Oxford) 2017; 56:1254-1263. [PMID: 27686101 DOI: 10.1093/rheumatology/kew318] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 01/18/2023] Open
Abstract
RA is the most common chronic systemic autoimmune disease, with a higher prevalence in women, suggesting female hormonal factors play a role in the development of the disease. However, many controversies still exist. The aim of this review was to appraise data from recent research concerning female hormonal factors and their association with RA disease development. The study of female hormonal factors is challenging because serum levels may differ throughout a woman's lifetime and interact with various environmental, immunological, genetic and endocrine factors influencing the development of autoimmunity. As some female hormonal factors may be potentially modifiable, understanding their impact on RA development is clinically relevant and may result in specific preventive interventions in high-risk populations.
Collapse
Affiliation(s)
| | - Nicola Pluchino
- Division of Gynecology, Department of Obstetrics and Gynecology, University Hospitals of Geneva, Geneva, Switzerland.,Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Geraldine Canny
- Department of Research affairs, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cem Gabay
- 3Division of Rheumatology, Department of Internal Medicine Specialties
| | - Axel Finckh
- 3Division of Rheumatology, Department of Internal Medicine Specialties
| |
Collapse
|
34
|
Falahee M, Simons G, Buckley CD, Hansson M, Stack RJ, Raza K. Patients' Perceptions of Their Relatives' Risk of Developing Rheumatoid Arthritis and of the Potential for Risk Communication, Prediction, and Modulation. Arthritis Care Res (Hoboken) 2017; 69:1558-1565. [DOI: 10.1002/acr.23179] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/17/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022]
Affiliation(s)
| | | | - Christopher D. Buckley
- University of Birmingham, and Sandwell and West Birmingham Hospitals NHS Trust; Birmingham UK
| | | | - Rebecca J. Stack
- University of Birmingham, Birmingham, and Trent University; Nottingham UK
| | - Karim Raza
- University of Birmingham, and Sandwell and West Birmingham Hospitals NHS Trust; Birmingham UK
| |
Collapse
|
35
|
Zou Y, Zhang Z, Liu Y, Liu D, Xu W. Are programmed cell death 1 gene polymorphisms correlated with susceptibility to rheumatoid arthritis?: A meta-analysis. Medicine (Baltimore) 2017; 96:e7805. [PMID: 28858091 PMCID: PMC5585485 DOI: 10.1097/md.0000000000007805] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Several studies investigated the relationship between programmed cell death 1 (PDCD1) gene polymorphisms and rheumatoid arthritis (RA) risk, but the results were controversial. To explore whether PDCD1 gene polymorphisms have an effect on RA risk, we conducted this meta-analysis to investigate the relationships between PDCD1 polymorphisms (rs36084323 [PD-1.1 G/A], rs11568821 [PD-1.3 G/A] and rs2227981 [PD-1.5 C/T]) and RA risk under 4 genetic models. METHODS PubMed, EMBASE, Web of Science, Cochrane Library China National Knowledge Infrastructure (CNKI), and Chinese Biomedical Literature Database (CBLM) were systematically searched for all eligible case-control studies. The last search was updated on September 10, 2016. Studies were accessed using Newcastle-Ottawa Scale case control study (NOS), and the combined effect size was calculated using STATA software, version 12.0. The pooled odds ratio (OR) with 95% confidence interval (CI) was calculated to assess the association. Heterogeneity analysis and subgroup analysis were also performed. Sensitivity analysis and publication bias were also performed if necessary. RESULTS This meta-analysis included 6 studies. The result demonstrated null association between rs36084323 (PD-1.1 G/A) polymorphism and RA susceptibility in all 4 genetic models. With regard to rs11568821 (PD-1.3 G/A), statistically significant association with RA risk was observed under allele model in Caucasians (allele model A vs G, OR = 1.19, 95% CI = 1.03-1.41). There was no significant association between rs2227981 (PD-1.5 C/T) polymorphism and RA risk. CONCLUSION The present study suggests that mutant A allele in rs11568821 (PD-1.3 G/A) might increase the susceptibility to RA in Caucasians.
Collapse
Affiliation(s)
- Yuming Zou
- Department of Orthopedics, Changhai Hospital, The First Affiliated Hospital of the Second Military Medical University
| | - Ziteng Zhang
- Department of Health Toxicology, College of Tropical Medicine and Public Health, Second Military Medical University, Shanghai, China
| | - Yangang Liu
- Department of Health Toxicology, College of Tropical Medicine and Public Health, Second Military Medical University, Shanghai, China
| | - Denghui Liu
- Department of Orthopedics, Changhai Hospital, The First Affiliated Hospital of the Second Military Medical University
| | - Weidong Xu
- Department of Orthopedics, Changhai Hospital, The First Affiliated Hospital of the Second Military Medical University
| |
Collapse
|
36
|
Yang A, Zhou J, Wang B, Dai J, Colman RW, Song W, Wu Y. A critical role for plasma kallikrein in the pathogenesis of autoantibody-induced arthritis. FASEB J 2017; 31:5419-5431. [PMID: 28808141 DOI: 10.1096/fj.201700018r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 07/31/2017] [Indexed: 01/08/2023]
Abstract
The plasma kallikrein-kinin system (KKS) consists of serine proteases, prekallikrein (pKal) and factor XII (FXII), and a cofactor, high-MW kininogen (HK). Upon activation, activated pKal and FXII cleave HK to release bradykinin. Activation of this system has been noted in patients with rheumatoid arthritis, and its pathogenic role has been characterized in animal arthritic models. In this study, we generated 2 knockout mouse strains that lacked pKal and HK and determined the role of KKS in autoantibody-induced arthritis. In a K/BxN serum transfer-induced arthritis (STIA) model, mice that lacked HK, pKal, or bradykinin receptors displayed protective phenotypes in joint swelling, histologic changes in inflammation, and cytokine production; however, FXII-deficient mice developed normal arthritis. Inhibition of Kal ameliorated arthritis severity and incidence at early stage STIA and reduced the levels of major cytokines in joints. In addition to releasing bradykinin from HK, Kal directly activated monocytes to produce proinflammatory cytokines, up-regulated their C5aR and FcRIII expression, and released C5a. Immune complex increased pKal activity, which led to HK cleavage. The absence of HK is associated with a decrease in joint vasopermeability. Thus, we identify a critical role for Kal in autoantibody-induced arthritis with pleiotropic effects, which suggests that it is a new target for the inhibition of arthritis.-Yang, A., Zhou, J., Wang, B., Dai, J., Colman, R. W., Song, W., Wu, Y. A critical role for plasma kallikrein in the pathogenesis of autoantibody-induced arthritis.
Collapse
Affiliation(s)
- Aizhen Yang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Junsong Zhou
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.,The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Bo Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jihong Dai
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, Rutgers-New Jersey Medical School, Newark, New Jersey, USA
| | - Robert W Colman
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Wenchao Song
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yi Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China; .,The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
37
|
Handa R, Upadhyaya S, Kapoor S, Jois R, Pandey BD, Bhatnagar AK, Khanna A, Goyal V, Kumar K. Tuberculosis and biologics in rheumatology: A special situation. Int J Rheum Dis 2017; 20:1313-1325. [PMID: 28730751 DOI: 10.1111/1756-185x.13129] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
India has a huge patient burden of rheumatic diseases (RDs) including rheumatoid arthritis. The use of biologics has transformed the treatment paradigm for RD; however, biologic treatment-related infections (especially tuberculosis [TB]) are an area of potential concern for TB-endemic nations like India. Anti-tumor necrosis factor (TNF) therapy impairs the physiological TNF-mediated signaling and may cause reactivation and dissemination of latent TB infection (LTBI). Careful screening is, thus, crucial in RD patients who are about to commence anti-TNF treatment. To date, there is no consensus available for the screening, evaluation and treatment of LTBI as well as on the drug dosage and duration regimen (monotherapy or combination therapy) in the Indian population. An evidence-based algorithm for LTBI screening and management in RD patients undergoing biologic disease-modifying anti-rheumatic drug therapy is suggested in this review for Indian rheumatologists. The proposed algorithm guides physicians through a step-wise screening approach, including medical history, tuberculin skin test, interferon gamma release assay, chest radiograph and management of LTBI with isoniazid therapy or its combination with rifampicin. Further, the provided algorithm can aid the national bodies (such as National TB Control Program) in formulating recommendations for LTBI in this high-risk population.
Collapse
Affiliation(s)
- Rohini Handa
- Department of Rheumatology, Indraprastha Apollo Hospital, New Delhi, India
| | - Sundeep Upadhyaya
- Department of Rheumatology, Indraprastha Apollo Hospital, New Delhi, India
| | - Sanjiv Kapoor
- Department of Rheumatology, Indian Spinal Injuries Center, New Delhi, India
| | - Ramesh Jois
- Department of Rheumatology, Fortis Hospital, Bangalore, India
| | | | - Anuj K Bhatnagar
- Rajan Babu Institute of Pulmonary Medicine and Tuberculosis, New Delhi, India
| | | | - Vishal Goyal
- Medical Affairs Department, Janssen India, Johnson & Johnson Pvt. Ltd., Mumbai, India
| | - Kamal Kumar
- Medical Affairs Department, Janssen India, Johnson & Johnson Pvt. Ltd., Mumbai, India
| |
Collapse
|
38
|
|
39
|
Intake of ω-3 polyunsaturated fatty acids in patients with rheumatoid arthritis: A systematic review and meta-analysis. Nutrition 2017; 45:114-124.e4. [PMID: 28965775 DOI: 10.1016/j.nut.2017.06.023] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/06/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic, autoimmune inflammatory disease of multiple joints that puts the patient at high risk for developing cardiovascular diseases (CVDs). The aim of the present study was to conduct an up-to-date systematic review and meta-analysis of published randomized controlled trials (RCTs) to assess potential changes in RA disease activity, inflammation, and CVD risk after oral intake of ω-3 polyunsaturated fatty acids. METHODS Publications up to July 31, 2016 were examined using the PubMed, SCOPUS, and EMBASE databases. INCLUSION CRITERIA English language; human subjects; both sexes; RCTs; oral intake of ω-3 fatty acids; minimum duration of 3 mo; and no medication change throughout intervention. The Cochrane Risk of Bias tool was used to assess quality of trials. We included 20 RCTs, involving 717 patients with RA in the intervention group and 535 RA patients in the control group. RESULTS Despite the evidence of overall low quality of trials, consumption of ω-3 fatty acids was found to significantly improve eight disease-activity-related markers. Regarding inflammation, only leukotriene B4 was reduced (five trials, standardized mean difference [SMD], -0.440; 95% confidence interval [CI], -0.676 to -0.205; I2 = 46.5%; P < 0.001). A significant amelioration was found for blood triacylglycerol levels (three trials, SMD, -0.316; 95% CI, -0.561 to -0.070; I2 = 0.0%; P = 0.012). CONCLUSION The beneficial properties of ω-3 polyunsaturated fatty acids on RA disease activity confirm the results of previous meta-analyses. Among five proinflammatory markers evaluated, only leukotriene B4 was found to be reduced. However, a positive effect on blood lipid profile of patients with RA was evident, perhaps for the first time.
Collapse
|
40
|
IgA N- and O-glycosylation profiling reveals no association with the pregnancy-related improvement in rheumatoid arthritis. Arthritis Res Ther 2017; 19:160. [PMID: 28679431 PMCID: PMC5498977 DOI: 10.1186/s13075-017-1367-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/09/2017] [Indexed: 02/01/2023] Open
Abstract
Background The Fc glycosylation of immunoglobulin G (IgG) is well known to associate with rheumatoid arthritis (RA) disease activity. The same may be true for other classes of Igs. In the present study, we sought to determine whether the glycosylation of IgA was different between healthy subjects and patients with RA, as well as whether it was associated with RA disease activity, in particular with the pregnancy-associated improvement thereof or the flare after delivery. Methods A recently developed high-throughput method for glycoprofiling of IgA1 was applied to affinity-captured IgA from sera of patients with RA (n = 252) and healthy control subjects (n = 32) collected before, during and after pregnancy. Results IgA1 O-glycans bore more sialic acids in patients with RA than in control subjects. In addition, levels of bisecting N-acetylglucosamine of the N-glycans at asparagine 144 were higher in the patients with RA. The levels of several N-glycosylation traits were shown to change with pregnancy, similar to what has been shown before for IgG. However, the changes in IgA glycosylation were not associated with improvement or a flare of disease activity. Conclusions The glycosylation of IgA differs between patients with RA and healthy control subjects. However, our data suggest only a minor, if any, association of IgA glycosylation with RA disease activity. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1367-0) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Koopman FA, van Maanen MA, Vervoordeldonk MJ, Tak PP. Balancing the autonomic nervous system to reduce inflammation in rheumatoid arthritis. J Intern Med 2017; 282:64-75. [PMID: 28547815 DOI: 10.1111/joim.12626] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Imbalance in the autonomic nervous system (ANS) has been observed in many established chronic autoimmune diseases, including rheumatoid arthritis (RA), which is a prototypic immune-mediated inflammatory disease (IMID). We recently discovered that autonomic dysfunction precedes and predicts arthritis development in subjects at risk of developing seropositive RA. In addition, RA patients with relatively high vagus nerve tone (higher parasympathetic parameters, measured by heart rate variability) respond better to antirheumatic therapies. Together, these data suggest that the ANS may control inflammation in humans. This notion is supported by experimental studies in animal models of RA. We have found that stimulation of the so-called cholinergic anti-inflammatory pathway by efferent electrical vagus nerve stimulation (VNS) or pharmacological activation of the alpha7 subunit of nicotinic acetylcholine receptors (α7nAChR) improves clinical signs and symptoms of arthritis, reduces cytokine production and protects against progressive joint destruction. Conversely, increased arthritis activity was observed in alpha7nAChR knockout mice. These studies together with previous work in animal models of sepsis and other forms of inflammation provided the rationale for an experimental clinical trial in patients with RA. We could for the first time show that an implantable vagus nerve stimulator inhibits peripheral blood cytokine production in humans. VNS significantly inhibited TNF and IL-6 production and improved RA disease severity, even in some patients with therapy-resistant disease. This work strongly supports further studies using a bioelectronic approach to treat RA and other IMIDs.
Collapse
Affiliation(s)
- F A Koopman
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - M A van Maanen
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - M J Vervoordeldonk
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Galvani Bioelectronics, Stevenage, UK
| | - P P Tak
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,GlaxoSmithKline, Stevenage, UK.,University of Cambridge, Cambridge, UK.,Ghent University, Ghent, Belgium
| |
Collapse
|
42
|
van Zanten A, Arends S, Roozendaal C, Limburg PC, Maas F, Trouw LA, Toes REM, Huizinga TWJ, Bootsma H, Brouwer E. Presence of anticitrullinated protein antibodies in a large population-based cohort from the Netherlands. Ann Rheum Dis 2017; 76:1184-1190. [PMID: 28043998 PMCID: PMC5530344 DOI: 10.1136/annrheumdis-2016-209991] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/18/2016] [Accepted: 11/20/2016] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To determine the prevalence of anticitrullinated protein antibodies (ACPAs) and their association with known rheumatoid arthritis (RA) risk factors in the general population. METHODS Lifelines is a multidisciplinary prospective population-based cohort study in the Netherlands. Cross-sectional data from 40 136 participants were used. The detection of ACPA was performed by measuring anti-CCP2 on the Phadia-250 analyser with levels ≥6.2 U/mL considered positive. An extensive questionnaire was taken on demographic and clinical information, including smoking, periodontal health and early symptoms of musculoskeletal disorders. RA was defined by a combination of self-reported RA, medication use for the indication of rheumatism and visiting a medical specialist within the last year. RESULTS Of the total 40 136 unselected individuals, 401 (1.0%) had ACPA level ≥6.2 U/mL. ACPA positivity was significantly associated with older age, female gender, smoking, joint complaints, RA and first degree relatives with rheumatism. Of the ACPA-positive participants, 22.4% had RA (15.2% had defined RA according to our criteria and 7.2% self-reported RA only). In participants without RA, 311 (0.8%) were ACPA-positive. In the non-RA group, older age, smoking and joint complaints remained significantly more frequently present in ACPA-positive compared with ACPA-negative participants. CONCLUSIONS In this large population-based study, the prevalence of ACPA levels ≥6.2 U/mL was 1.0% for the total group and 0.8% when excluding patients with RA. Older age, smoking and joint complaints were more frequently present in ACPA-positive Lifelines participants. To our knowledge, this study is the largest study to date on ACPA positivity in the general, mostly Caucasian population.
Collapse
Affiliation(s)
- A van Zanten
- Department of Rheumatology and Clinical Immunology, Groningen, The Netherlands
| | - S Arends
- Department of Rheumatology and Clinical Immunology, Groningen, The Netherlands
| | - C Roozendaal
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - P C Limburg
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - F Maas
- Department of Rheumatology and Clinical Immunology, Groningen, The Netherlands
| | - L A Trouw
- Department of Rheumatology, LUMC, Leiden, The Netherlands
| | - R E M Toes
- Department of Rheumatology, LUMC, Leiden, The Netherlands
| | - T W J Huizinga
- Department of Rheumatology, LUMC, Leiden, The Netherlands
| | - H Bootsma
- Department of Rheumatology and Clinical Immunology, Groningen, The Netherlands
| | - E Brouwer
- Department of Rheumatology and Clinical Immunology, Groningen, The Netherlands
| |
Collapse
|
43
|
Alpizar-Rodriguez D, Mueller RB, Möller B, Dudler J, Ciurea A, Zufferey P, Kyburz D, Walker UA, von Mühlenen I, Roux-Lombard P, Mahler M, Lamacchia C, Courvoisier DS, Gabay C, Finckh A. Female hormonal factors and the development of anti-citrullinated protein antibodies in women at risk of rheumatoid arthritis. Rheumatology (Oxford) 2017; 56:1579-1585. [DOI: 10.1093/rheumatology/kex239] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 12/13/2022] Open
|
44
|
Burmester GR, Pope JE. Novel treatment strategies in rheumatoid arthritis. Lancet 2017; 389:2338-2348. [PMID: 28612748 DOI: 10.1016/s0140-6736(17)31491-5] [Citation(s) in RCA: 686] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/27/2017] [Accepted: 05/03/2017] [Indexed: 02/06/2023]
Abstract
New treatment strategies have substantially changed the course of rheumatoid arthritis. Many patients can achieve remission if the disease is recognised early and is treated promptly and continuously; however, some individuals do not respond adequately to treatment. Rapid diagnosis and a treat-to-target approach with tight monitoring and control, can increase the likelihood of remission in patients with rheumatoid arthritis. In this Series paper, we describe new insights into the management of rheumatoid arthritis with targeted therapy approaches using classic and novel medications, and outline the potential effects of precision medicine in this challenging disease. Articles are included that investigate the treat-to-target approach, which includes adding or de-escalating treatment. Rheumatoid arthritis treatment is impeded by delayed diagnosis, problematic access to specialists, and difficulties adhering to treat-to-target principles. Clinical management goals in rheumatoid arthritis include enabling rapid access to optimum diagnosis and care and the well informed use of multiple treatments approved for this disease.
Collapse
Affiliation(s)
- Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité-University Medicine Berlin, Berlin, Germany.
| | - Janet E Pope
- Division of Rheumatology, St Joseph's Hospital, Western University, London, ON, Canada
| |
Collapse
|
45
|
McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet 2017; 389:2328-2337. [PMID: 28612747 DOI: 10.1016/s0140-6736(17)31472-1] [Citation(s) in RCA: 901] [Impact Index Per Article: 112.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis is a chronic autoimmune disease that causes progressive articular damage, functional loss, and comorbidity. The development of effective biologics and small-molecule kinase inhibitors in the past two decades has substantially improved clinical outcomes. Just as understanding of pathogenesis has led in large part to the development of drugs, so have mode-of-action studies of these specific immune-targeted agents revealed which immune pathways drive articular inflammation and related comorbidities. Cytokine inhibitors have definitively proven a critical role for tumour necrosis factor α and interleukin 6 in disease pathogenesis and possibly also for granulocyte-macrophage colony-stimulating factor. More recently, clinical trials with Janus kinase (JAK) inhibitors have shown that cytokine receptors that signal through the JAK/STAT signalling pathway are important for disease, informing the pathogenetic function of additional cytokines (such as the interferons). Finally, successful use of costimulatory blockade and B-cell depletion in the clinic has revealed that the adaptive immune response and the downstream events initiated by these cells participate directly in synovial inflammation. Taken together, it becomes apparent that understanding the effects of specific immune interventions can elucidate definitive molecular or cellular nodes that are essential to maintain complex inflammatory networks that subserve diseases like rheumatoid arthritis.
Collapse
Affiliation(s)
- Iain B McInnes
- Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany; Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
46
|
Deane KD, Striebich CC, Holers VM. Editorial: Prevention of Rheumatoid Arthritis: Now Is the Time, but How to Proceed? Arthritis Rheumatol 2017; 69:873-877. [PMID: 28217918 DOI: 10.1002/art.40061] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
|
47
|
Abstract
One of the fundamental traits of immune cells in rheumatoid arthritis (RA) is their ability to proliferate, a property shared with the joint-resident cells that form the synovial pannus. The building of biomass imposes high demands for energy and biosynthetic precursors, implicating metabolic control as a basic disease mechanism. During preclinical RA, when autoreactive T cells expand and immunological tolerance is broken, the main sites of disease are the secondary lymphoid tissues. Naive CD4+ T cells from patients with RA have a distinct metabolic signature, characterized by dampened glycolysis, low ATP levels and enhanced shunting of glucose into the pentose phosphate pathway. Equipped with high levels of NADPH and depleted of intracellular reactive oxygen species, such T cells hyperproliferate and acquire proinflammatory effector functions. During clinical RA, immune cells coexist with stromal cells in the acidic milieu of the inflamed joint. This microenvironment is rich in metabolic intermediates that are released into the extracellular space to shape cell-cell communication and the functional activity of tissue-resident cells. Increasing awareness of how metabolites regulate signalling pathways, guide post-translational modifications and condition the tissue microenvironment will help to connect environmental factors with the pathogenic behaviour of T cells in RA.
Collapse
|
48
|
Deane KD, Demoruelle MK, Kelmenson LB, Kuhn KA, Norris JM, Holers VM. Genetic and environmental risk factors for rheumatoid arthritis. Best Pract Res Clin Rheumatol 2017; 31:3-18. [PMID: 29221595 PMCID: PMC5726551 DOI: 10.1016/j.berh.2017.08.003] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/12/2017] [Accepted: 07/17/2017] [Indexed: 01/13/2023]
Abstract
Multiple genetic and environmental factors have been associated with an increased risk for rheumatoid arthritis (RA). Of these, the strongest associations have been seen with female sex, a family history of RA, the genetic factor the "shared epitope," and exposure to tobacco smoke. There is also renewed interest in mucosal inflammation and microbial factors as contributors to the development of RA. However, the identification of a "preclinical" period of RA that can be defined as local or systemic autoimmunity as measured by autoantibodies and other biomarkers prior to the development of clinically apparent synovitis suggests that the risk factors for RA are acting long prior to first clinical evidence of IA. As such, a major challenge to the field will be to investigate the full spectrum of the development of RA, from initiation and propagation of autoimmunity during preclinical RA and transition to clinically apparent synovitis and classifiable RA, to determine which genetic and environmental factors are important at each stage of disease development. Understanding the exact role and timing of action of risk factors for RA is especially important given the advent of prevention trials in RA, and the hope that a full understanding of genetic and environmental factors in RA could lead to effective preventive interventions.
Collapse
Affiliation(s)
- Kevin D Deane
- Division of Rheumatology, University of Colorado Denver School of Medicine, USA.
| | | | - Lindsay B Kelmenson
- Division of Rheumatology, University of Colorado Denver School of Medicine, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado Denver School of Medicine, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, USA
| | - V Michael Holers
- Division of Rheumatology, University of Colorado Denver School of Medicine, USA
| |
Collapse
|
49
|
Eriksson K, Lönnblom E, Tour G, Kats A, Mydel P, Georgsson P, Hultgren C, Kharlamova N, Norin U, Jönsson J, Lundmark A, Hellvard A, Lundberg K, Jansson L, Holmdahl R, Yucel-Lindberg T. Effects by periodontitis on pristane-induced arthritis in rats. J Transl Med 2016; 14:311. [PMID: 27809921 PMCID: PMC5094068 DOI: 10.1186/s12967-016-1067-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/20/2016] [Indexed: 12/29/2022] Open
Abstract
Background An infection-immune association of periodontal disease with rheumatoid arthritis has been suggested. This study aimed to investigate the effect of pre-existing periodontitis on the development and the immune/inflammatory response of pristane-induced arthritis. Methods We investigated the effect of periodontitis induced by ligature placement and Porphyromonas gingivalis (P. gingivalis) infection, in combination with Fusobacterium nucleatum to promote its colonization, on the development of pristane-induced arthritis (PIA) in rats (Dark Agouti). Disease progression and severity of periodontitis and arthritis was monitored using clinical assessment, micro-computed tomography (micro-CT)/intraoral radiographs, antibody response, the inflammatory markers such as α-1-acid glycoprotein (α-1-AGP) and c-reactive protein (CRP) as well as cytokine multiplex profiling at different time intervals after induction. Results Experimentally induced periodontitis manifested clinically (P < 0.05) prior to pristane injection and progressed steadily until the end of experiments (15 weeks), as compared to the non-ligated arthritis group. Injection of pristane 8 weeks after periodontitis-induction led to severe arthritis in all rats demonstrating that the severity of arthritis was not affected by the pre-existence of periodontitis. Endpoint analysis showed that 89% of the periodontitis-affected animals were positive for antibodies against arginine gingipain B and furthermore, the plasma antibody levels to a citrullinated P. gingivalis peptidylarginine deiminase (PPAD) peptide (denoted CPP3) were significantly (P < 0.05) higher in periodontitis rats with PIA. Additionally, there was a trend towards increased pro-inflammatory and anti-inflammatory cytokine levels, and increased α-1-AGP levels in plasma from periodontitis-challenged PIA rats. Conclusions Pre-existence of periodontitis induced antibodies against citrullinated peptide derived from PPAD in rats with PIA. However, there were no differences in the development or severity of PIA between periodontitis challenged and periodontitis free rats. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1067-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaja Eriksson
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden.
| | - Erik Lönnblom
- Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Gregory Tour
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden.,Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Alfred Nobels Allé 8, 141 83, Huddinge, Sweden
| | - Anna Kats
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden
| | - Piotr Mydel
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, The Laboratory Building, 5th Floor, 5021, Bergen, Norway
| | - Pierre Georgsson
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden
| | - Catharina Hultgren
- Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Alfred Nobels Allé 8, 141 83, Huddinge, Sweden
| | - Nastya Kharlamova
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden
| | - Ulrika Norin
- Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jörgen Jönsson
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden
| | - Anna Lundmark
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden
| | - Annelie Hellvard
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, The Laboratory Building, 5th Floor, 5021, Bergen, Norway.,Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387, Kraków, Poland
| | - Karin Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Rheumatology Clinic D2:01, 171 76, Stockholm, Sweden
| | - Leif Jansson
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden.,Department of Periodontology at Eastmaninstitutet, Stockholm County Council, Dalagatan 11, 113 24, Stockholm, Sweden
| | - Rikard Holmdahl
- Section for Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.,Center for Medical Immunopharmacology Research, Southern Medical University, Guangzhou, China
| | - Tülay Yucel-Lindberg
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Box 4064, 141 04, Huddinge, Sweden.
| |
Collapse
|
50
|
Tang MW, Koopman FA, Visscher JPM, de Hair MJ, Gerlag DM, Tak PP. Hormone, metabolic peptide, and nutrient levels in the earliest phases of rheumatoid arthritis-contribution of free fatty acids to an increased cardiovascular risk during very early disease. Clin Rheumatol 2016; 36:269-278. [PMID: 27807638 PMCID: PMC5290053 DOI: 10.1007/s10067-016-3456-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/16/2016] [Indexed: 12/21/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease associated with changes in several hormones and metabolic peptides. Crosstalk between these factors and the immune system may be important for homeostasis during inflammation. Here, we studied the levels of hormones, metabolic peptides, and nutrients in individuals at risk for developing RA (at risk). In total, 18 hormones, metabolic peptides, and nutrients were measured in fasting serum samples from 45 autoantibody-positive individuals at risk, 22 RA patients, and 16 healthy subjects. Triglyceride (TG) levels were also measured in an independent validation cohort of 32 individuals at risk, 20 early arthritis patients, and 20 healthy controls. We found an elevated TG level in individuals at risk and significantly higher TG levels in RA patients compared to healthy controls. These results were confirmed in the validation cohort. Similarly, free fatty acid (FFA) levels showed an increase in individuals at risk and were significantly higher in RA patients compared to healthy controls. In RA patients, FFA levels were positively correlated with disease activity. Pancreatic polypeptide (PP) and norepinephrine levels were highly significantly increased in individuals at risk and RA patients compared to healthy controls. TG and FFA levels are increased in RA patients and positively correlated with disease activity parameters. The results presented here suggest a role for FFAs in the pathogenesis of RA. Furthermore, PP and norepinephrine may be a biomarker that could assist in the identification of individuals at risk.
Collapse
Affiliation(s)
- Man Wai Tang
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Frieda A Koopman
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Jan P M Visscher
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Maria J de Hair
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Danielle M Gerlag
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands.,Currently also Clinical Unit Cambridge, GlaxoSmithKline, Cambridge, UK
| | - Paul Peter Tak
- Department of Clinical Immunology and Rheumatology, Amsterdam Rheumatology and Immunology Center, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands. .,Currently also GlaxoSmithKline, Stevenage, UK. .,University of Cambridge, Cambridge, UK. .,Ghent University, Ghent, Belgium.
| |
Collapse
|