1
|
Wei S, Cheng RJ, Li S, Lu C, Zhang Q, Wu Q, Zhao X, Tian X, Zeng X, Liu Y. MSC-microvesicles protect cartilage from degradation in early rheumatoid arthritis via immunoregulation. J Nanobiotechnology 2024; 22:673. [PMID: 39497131 PMCID: PMC11536868 DOI: 10.1186/s12951-024-02922-6] [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: 05/26/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
OBJECTIVE As research into preclinical rheumatoid arthritis (pre-RA) has advanced, a growing body of evidence suggests that abnormalities in RA-affected joint cartilage precede the onset of arthritis. Thus, early prevention and treatment strategies are imperative. In this study, we aimed to explore the protective effects of mesenchymal stem cell (MSC)-derived microvesicles (MVs) on cartilage degradation in a collagen-induced arthritis (CIA) mouse model. METHODS A CIA mouse model was established to observe early pathological changes in cartilage (days 21-25) through histological and radiological examinations. On day 22, MSCs-MVs were intravenously injected into the mice with CIA. Radiological, histological, and flow cytometric examinations were conducted to observe inflammation and cartilage changes in these mice compared to the mice with CIA and the control mice. In vitro, chondrocytes were cultured with inflammatory factors such as IL-1β and TNFα to simulate inflammatory damage to cartilage. After the addition of MVs, changes in inflammatory levels and collagen expression were measured via Western blotting, immunofluorescence, enzyme-linked immunosorbent assays (ELISAs), and quantitative PCR to determine the role of MVs in maintaining chondrocytes. RESULTS MSC-MVs expressed vesicular membrane proteins (CD63 and Annexin V) and surface markers characteristic of MSCs (CD44, CD73, CD90, and CD105). In the early stages of CIA in mice, a notable decrease in collagen content was observed in the joint cartilage. In mice with CIA, injection of MSCs-MVs resulted in a significant reduction in the peripheral blood levels of IL-1β, TNFα, and IL-6, along with a decrease in the ratio of proinflammatory T and B cells. Additionally, MSC-MVs downregulated the expression of IL-1β, TNFα, MMP-13, and ADAMTS-5 in cartilage while maintaining the stability of type I and type II collagen. These MVs also attenuated the destruction of cartilage, which was evident on imaging. In vitro experiments demonstrated that MSC-MVs effectively suppressed the secretion of the inflammatory factors IL-1β, TNFα, and IL-6 in stimulated peripheral blood mononuclear cells (PBMCs). CONCLUSIONS MSCs-MVs can inhibit the decomposition of the inflammation-induced cartilage matrix by regulating immune cell inflammatory factors to attenuate cartilage destruction. MSC-MVs are promising effective treatments for the early stages of RA.
Collapse
Affiliation(s)
- Shixiong Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Sujia Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenyang Lu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiuhong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xueting Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Cîrciumaru A, Kisten Y, Hansson M, Mathsson-Alm L, Joshua V, Wähämaa H, Loberg Haarhaus M, Lindqvist J, Padyukov L, Catrina SB, Fei G, Vivar N, Rezaei H, af Klint E, Antovic A, Réthi B, Catrina AI, Hensvold A. Identification of early risk factors for anti-citrullinated-protein-antibody positive rheumatoid arthritis-a prospective cohort study. Rheumatology (Oxford) 2024; 63:3164-3171. [PMID: 38457608 PMCID: PMC11534094 DOI: 10.1093/rheumatology/keae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/16/2024] [Accepted: 02/09/2024] [Indexed: 03/10/2024] Open
Abstract
OBJECTIVE Individuals positive for anti-cyclic-peptide-antibodies (anti-CCP) and musculoskeletal complaints (MSK-C) are at risk for developing rheumatoid arthritis (RA). In this study we aimed to investigate factors involved in arthritis progression. METHODS Anti-CCP2-positive individuals with MSK-C referred to a rheumatologist were recruited. Individuals lacked arthritis at clinical and ultrasound examination and were followed for ≥3 years or until clinical arthritis diagnosis. Blood samples from inclusion were analysed for nine ACPA reactivities (citrullinated α-1-enolase, fibrinogen, filaggrin, histone, vimentin and tenascin peptides); 92 inflammation-associated proteins; and HLA-shared epitope alleles. Cox regression was applied to the data to identify independent predictors in a model. RESULTS Two hundred and sixty-seven individuals were included with median follow-up of 49 months (interquartile range [IQR]: 22-60); 101 (38%) developed arthritis after a median of 14 months (IQR: 6-27). The analysis identified that presence of at least one ACPA reactivity (hazard ratio [HR] 8.0; 95% CI: 2.9, 22), ultrasound-detected tenosynovitis (HR 3.4; 95% CI: 2.0, 6.0), IL-6 levels (HR 1.5; 95% CI: 1.2, 1.8) and IL-15 receptor α (IL-15Rα) levels (HR 0.6; 95% CI: 0.4, 0.9) are significant independent predictors for arthritis progression in a prediction model (Harrell's C 0.76 [s.e. 0.02], AUC 0.82 [95% CI: 0.76, 0.89], cross-validated AUC 0.70 [95% CI: 0.56, 0.85]). CONCLUSION We propose a high RA risk phase characterized by presence of ACPA reactivity, tenosynovitis, IL-6 and IL-15Rα and suggest that these factors need to be further investigated for their biological effects and clinical values, to identify individuals at particular low risk and high risk for arthritis progression.
Collapse
Affiliation(s)
- Alexandra Cîrciumaru
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm
| | - Yogan Kisten
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Monika Hansson
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Thermo Fisher Scientific, Uppsala, Sweden
| | | | - Vijay Joshua
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Heidi Wähämaa
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Malena Loberg Haarhaus
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Rheumatology, Karolinska University Hospital
| | - Joakim Lindqvist
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Rheumatology, Karolinska University Hospital
| | - Leonid Padyukov
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Diabetes, Academic Specialist Centrum, Stockholm, Sweden
| | - Guozhong Fei
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm
- Swedish Medical Products Agency, Uppsala, Sweden
| | - Nancy Vivar
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Rheumatology, Karolinska University Hospital
| | - Hamed Rezaei
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Rheumatology, Karolinska University Hospital
| | - Erik af Klint
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Rheumatology, Karolinska University Hospital
| | - Aleksandra Antovic
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm
- Department of Rheumatology, Karolinska University Hospital
| | - Bence Réthi
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anca I Catrina
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm
- Department of Rheumatology, Karolinska University Hospital
| | - Aase Hensvold
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm Health Services, Region Stockholm
| |
Collapse
|
3
|
Fautrel B, Kedra J, Rempenault C, Juge PA, Drouet J, Avouac J, Baillet A, Brocq O, Alegria GC, Constantin A, Dernis E, Gaujoux-Viala C, Goëb V, Gottenberg JE, Le Goff B, Marotte H, Richez C, Salmon JH, Saraux A, Senbel E, Seror R, Tournadre A, Vittecoq O, Escaffre P, Vacher D, Dieudé P, Daien C. 2024 update of the recommendations of the French Society of Rheumatology for the diagnosis and management of patients with rheumatoid arthritis. Joint Bone Spine 2024; 91:105790. [PMID: 39389412 DOI: 10.1016/j.jbspin.2024.105790] [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: 07/15/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
The French Society of Rheumatology recommendations for managing rheumatoid arthritis (RA) has been updated by a working group of 21 rheumatology experts, 4 young rheumatologists and 2 patient association representatives on the basis of the 2023 version of the European Alliance of Associations for Rheumatology (EULAR) recommendations and systematic literature reviews. Two additional topics were addressed: people at risk of RA development and RA-related interstitial lung disease (RA-ILD). Four general principles and 19 recommendations were issued. The general principles emphasize the importance of a shared decision between the rheumatologist and patient and the need for comprehensive management, both drug and non-drug, for people with RA or at risk of RA development. In terms of diagnosis, the recommendations stress the importance of clinical arthritis and in its absence, the risk factors for progression to RA. In terms of treatment, the recommendations incorporate recent data on the cardiovascular and neoplastic risk profile of Janus kinase inhibitors. With regard to RA-ILD, the recommendations highlight the importance of clinical screening and the need for high-resolution CT scan in the presence of pulmonary symptoms. RA-ILD management requires collaboration between rheumatologists and pulmonologists. The treatment strategy is based on controlling disease activity with methotrexate or targeted therapies (mainly abatacept or rituximab). The prescription for anti-fibrotic treatment should be discussed with a pulmonologist with expertise in RA-ILD.
Collapse
Affiliation(s)
- Bruno Fautrel
- Sorbonne université, Paris, France; Service de rhumatologie, groupe hospitalier Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris, 75013 Paris, France; Inserm UMRS 1136, PEPITES Team, 75013 Paris, France; CRI-IMIDIATE Clinical Research Network, 75013 Paris, France.
| | - Joanna Kedra
- Sorbonne université, Paris, France; Service de rhumatologie, groupe hospitalier Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris, 75013 Paris, France; Inserm UMRS 1136, PEPITES Team, 75013 Paris, France; CRI-IMIDIATE Clinical Research Network, 75013 Paris, France
| | - Claire Rempenault
- Université Paris-Cité, Paris, France; Service de rhumatologie, groupe hospitalier Bichat - Claude-Bernard, Assistance publique-Hôpitaux de Paris, 75018 Paris, France
| | - Pierre-Antoine Juge
- Inserm UMRS 1152, équipe 2, 75018 Paris, France; Université de Montpellier, Montpellier, France; Service de rhumatologie, CHU de Montpellier, CHU Lapeyronie, Montpellier, France
| | | | - Jérôme Avouac
- Department of Rheumatology, Hôpital Cochin, AP-HP, Paris, France; Université Paris-Cité, Paris, France; Inserm U1016, UMR 8104, Paris, France
| | - Athan Baillet
- TIMC, UMR 5525, university Grenoble-Alpes, Grenoble, France
| | - Olivier Brocq
- Rheumatology, Princess-Grace Hospital, boulevard Pasteur, 98000 Monaco, Monaco
| | - Guillermo Carvajal Alegria
- Service de rhumatologie, hôpital Trousseau, CHRU de Tours, 37044 Tours cedex, France; UFR Medicine, University of Tours, Tours, France; UPR 4301 CNRS Centre de Biophysique Moléculaire, Nanomedicaments et Nanosondes Department, Tours, France
| | - Arnaud Constantin
- Service de rhumatologie, hôpital Pierre-Paul-Riquet, CHU de Purpan, Toulouse, France; Université de Toulouse III - Paul-Sabatier, Toulouse, France; INFINITY, Inserm UMR 1291, CHU de Purpan, Toulouse, France
| | | | - Cécile Gaujoux-Viala
- Inserm, IDESP, University of Montpellier, Montpellier, France; Rheumatology Department, CHU of Nîmes, Nîmes, France
| | - Vincent Goëb
- Rheumatology, Autonomy Unit, UPJV, CHU of Amiens-Picardie, 80000 Amiens, France
| | | | - Benoit Le Goff
- Rheumatology Department, CHU of Nantes, 44000 Nantes, France
| | - Hubert Marotte
- Rheumatology Department, Université Jean-Monnet Saint-Étienne, Saint-Étienne, France; Inserm, SAINBIOSE U1059, Mines Saint-Étienne, CHU of Saint-Etienne, 42023 Saint-Étienne, France
| | - Christophe Richez
- Service de rhumatologue, centre national de référence des maladies auto-immunes systémiques rares RESO, Bordeaux, France; UMR/CNRS 5164, ImmunoConcEpT, CNRS, hôpital Pellegrin, université de Bordeaux, CHU de Bordeaux, Bordeaux, France
| | | | - Alain Saraux
- Université de Bretagne-Occidentale, université de Brest, Brest, France; Inserm (U1227), LabEx IGO, Department of Rheumatology, CHU of Brest, 29200 Brest, France
| | - Eric Senbel
- Conseil National Professionnel de Rhumatologie, France
| | - Raphaèle Seror
- Department of Rheumatology, Hôpital Bicêtre, AP-HP, Paris, France; Inserm-UMR 1184, centre national de référence des maladies auto-immunes systémiques rares, université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Anne Tournadre
- UNH INRAe UCA, Rheumatology Department, CHU of Clermont-Ferrand, Clermont-Ferrand, France
| | | | | | | | - Philippe Dieudé
- Inserm UMRS 1152, équipe 2, 75018 Paris, France; Service de rhumatologie, groupe hospitalier Bichat, université de Paris, Assistance publique-Hôpitaux de Paris, 75018 Paris, France
| | - Claire Daien
- Université de Montpellier, Montpellier, France; Service de rhumatologie, CHU de Montpellier, CHU Lapeyronie, Montpellier, France; Inserm U1046, CNRS UMR 9214, University of Montpellier, Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), Montpellier, France
| |
Collapse
|
4
|
Mansouri P, Mansouri P, Behmard E, Najafipour S, Kouhpayeh SA, Farjadfar A. Peptidylarginine deiminase (PAD): A promising target for chronic diseases treatment. Int J Biol Macromol 2024; 278:134576. [PMID: 39127273 DOI: 10.1016/j.ijbiomac.2024.134576] [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: 05/30/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
In 1958, the presence of citrulline in the structure of the proteins was discovered for the first time. Several years later they found that Arginine converted to citrulline during a post-translational modification process by PAD enzyme. Each PAD is expressed in a certain tissue developing a series of diseases such as inflammation and cancers. Among these, PAD2 and PAD4 play a role in the development of rheumatoid arthritis (RA) by producing citrullinated autoantigens and increasing the production of inflammatory cytokines. PAD4 is also associated with the formation of NET structures and thrombosis. In the crystallographic structure, PAD has several calcium binding sites, and the active site of the enzyme consists of different amino acids. Various PAD inhibitors have been developed divided into pan-PAD and selective PAD inhibitors. F-amidine, Cl-amidine, and BB-Cl-amidine are some of pan-PAD inhibitors. AFM-30a and JBI589 are selective for PAD2 and PAD4, respectively. There is a need to evaluate the effectiveness of existing inhibitors more accurately in the coming years, as well as design and production of novel inhibitors targeting highly specific isoforms.
Collapse
Affiliation(s)
- Pegah Mansouri
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Pardis Mansouri
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Sohrab Najafipour
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Seyed Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Akbar Farjadfar
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
5
|
Hinkema HJ, Westra J, Arends S, Brouwer E, Mulder DJ. Higher levels of markers for early atherosclerosis in anti-citrullinated protein antibodies positive individuals at risk for RA, a cross sectional study. Rheumatol Int 2024; 44:2007-2016. [PMID: 39012360 PMCID: PMC11393035 DOI: 10.1007/s00296-024-05659-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
OBJECTIVE To identify differences in levels of serum biomarkers associated with atherosclerosis between anti-citrullinated protein antibodies (ACPA) positive groups. METHODS Cross-sectional data were used from the Dutch Lifelines Cohort Study combined with data derived from RA risk and early RA studies conducted at the University Medical Center Groningen (UMCG). Serum biomarkers of inflammation, endothelial cell activation, tissue remodeling and adipokine, which were previously associated with atherosclerosis, were measured with Luminex in four ACPA positive groups with different characteristics: without joint complaints, with joint complaints, RA risk and early RA groups. RESULTS Levels of C-reactive protein (CRP), Interleukin-6 (IL-6), Tumor Necrosis Factor Receptor 1 (TNFR1) and vascular endothelial growth factor (VEGF) were significantly higher in the RA risk and early RA groups compared to the joint complaints and the no joint complaints groups. The difference remained statistically significant after correcting for renal function, smoking and hypertension in multivariate logistic regression analysis, with focus on ACPA positive with joint complaints group versus RA risk group: CRP OR = 2.67, p = 0.033; IL-6 OR = 3.73, p = 0.019; TNFR1 OR = 1.003, p < 0.001; VGEF OR = 8.59, p = 0.019. CONCLUSION Individuals at risk for RA have higher levels of inflammatory markers and VEGF, which suggests that they might also have a risk of higher cardiovascular disease (CVD); however, this does not apply to individuals with ACPA positivity with self-reported joint complaints or without joint complaints only. Therefore, it is important that individuals with RA risk are referred to a rheumatologist to rule in or out arthritis/development of RA and discuss CVD risk.
Collapse
Affiliation(s)
- Helma J Hinkema
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, P.O. Box 30001, Groningen, 9700 RB, The Netherlands.
| | - Johanna Westra
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, P.O. Box 30001, Groningen, 9700 RB, The Netherlands
| | - Suzanne Arends
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, P.O. Box 30001, Groningen, 9700 RB, The Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, P.O. Box 30001, Groningen, 9700 RB, The Netherlands
| | - Douwe J Mulder
- Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Holers VM, Demoruelle KM, Buckner JH, James EA, Firestein GS, Robinson WH, Steere AC, Zhang F, Norris JM, Kuhn KA, Deane KD. Distinct mucosal endotypes as initiators and drivers of rheumatoid arthritis. Nat Rev Rheumatol 2024; 20:601-613. [PMID: 39251771 DOI: 10.1038/s41584-024-01154-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Rheumatoid arthritis (RA) is a potentially devastating autoimmune disease. The great majority of patients with RA are seropositive for anti-citrullinated protein antibodies (ACPAs), rheumatoid factors, or other autoantibodies. The onset of clinically apparent inflammatory arthritis meeting classification criteria (clinical RA) is preceded by ACPA seropositivity for an average of 3-5 years, a period that is designated as 'at-risk' of RA for ACPA-positive individuals who do not display signs of arthritis, or 'pre-RA' for individuals who are known to have progressed to developing clinical RA. Prior studies of individuals at-risk of RA have associated pulmonary mucosal inflammation with local production of ACPAs and rheumatoid factors, leading to development of the 'mucosal origins hypothesis'. Recent work now suggests the presence of multiple distinct mucosal site-specific mechanisms that drive RA evolution. Indicatively, subsets of individuals at-risk of RA and patients with RA harbour a faecal bacterial strain that has exhibited arthritogenic activity in animal models and that favours T helper 17 (TH17) cell responses in patients. Periodontal inflammation and oral microbiota have also been suggested to promote the development of arthritis through breaches in the mucosal barrier. Herein, we argue that mucosal sites and their associated microbial strains can contribute to RA evolution via distinct pathogenic mechanisms, which can be considered causal mucosal endotypes. Future therapies instituted for prevention in the at-risk period, or, perhaps, during clinical RA as therapeutics for active arthritis, will possibly have to address these individual mechanisms as part of precision medicine approaches.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA.
| | | | | | | | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, CA, USA
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Fan Zhang
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
7
|
Goff SH, Bergstedt DT, Feser ML, Moss L, Mikuls TR, Edison JD, Holers VM, Martinez‐Prat L, Aure MAR, Mahler M, Deane KD. Multi-Autoantibody Testing Identifies Expansion of Reactivity to Targeted Antigens Before a Diagnosis of Rheumatoid Arthritis. ACR Open Rheumatol 2024; 6:587-597. [PMID: 38950890 PMCID: PMC11506557 DOI: 10.1002/acr2.11704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 07/03/2024] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) has a "pre-RA" period in which multiple autoantibodies, including antibodies to citrullinated (cit) proteins (ACPA), rheumatoid factor (RF), anti-peptidyl arginine deiminase (anti-PAD), among others, have been described; however, few studies have tested all autoantibodies in a single pre-RA cohort. This study aims to evaluate the prevalence of multiple autoantibodies in pre-RA and potentially identify an autoantibody profile in pre-RA that indicates imminent onset of clinical RA. METHODS We evaluated 148 individuals with two pre- and one post-RA diagnosis samples available from the Department of Defense Serum Repository and matched controls. Samples were tested for immuglobulin (Ig) G anti-cyclic cit peptide-3 (anti-CCP3), five ACPA fine specificities, five anti-PAD isoforms, as well as RF IgA and RF IgM using commercial platforms; cutoffs were determined using levels present in <1% of controls. RESULTS Positivity of anti-CCP3, RF IgA and RF IgM, anti-PAD1, anti-cit-vimentin 2, anti-cit-fibrinogen, and anti-cit-histone 1 increased over time in pre-RA, although anti-PAD and ACPA fine specificities were predominately present within anti-CCP3-positive individuals. Within anti-CCP3-positive samples from the pre-RA period, positivity for RFs as well as anti-PAD and ACPA fine specificities classified samples as being closer to the time of RA diagnosis. CONCLUSION Multiple autoantibodies are present in pre-RA and increase in positivity as the time of RA diagnosis approaches. These results confirm previous findings predicting imminent RA and provide a pathway using commercial-grade assays to assess the risk for and timing of development of clinical RA.
Collapse
Affiliation(s)
| | | | | | - LauraKay Moss
- University of Colorado Anschutz Medical CampusAurora
| | - Ted R. Mikuls
- University of Nebraska Medical Center and VA Nebraska‐Western Iowa Health Care SystemOmaha
| | - Jess D. Edison
- Walter Reed National Military Medical Center and Uniformed Services University of the Health SciencesBethesdaMaryland
| | | | | | | | | | | |
Collapse
|
8
|
Aripova N, Thiele GM, Duryee MJ, Hunter CD, Yang Y, Roul P, Ascherman DP, Matson SM, Kunkel G, Cannon GW, Wysham KD, Kerr GS, Monach PA, Baker JF, Poole JA, Mikuls TR, England BR. Antibodies to Malondialdehyde-Acetaldehyde Adduct Are Associated With Prevalent and Incident Rheumatoid Arthritis-Associated Interstitial Lung Disease in US Veterans. Arthritis Rheumatol 2024; 76:1353-1363. [PMID: 38766737 PMCID: PMC11349468 DOI: 10.1002/art.42916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/15/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVE The objective of this study is to determine the associations of protein-specific anti-malondialdehyde-acetaldehyde (MAA) antibodies with prevalent and incident rheumatoid arthritis-interstitial lung disease (RA-ILD). METHODS Within a multicenter, prospective cohort of US veterans with RA, RA-ILD was validated by medical record review of clinical diagnoses, chest imaging, and pathology. Serum antibodies to MAA-albumin, MAA-collagen, MAA-fibrinogen, and MAA-vimentin (IgA, IgM, and IgG) were measured by a standardized enzyme-linked immunosorbent assay. Associations of anti-MAA antibodies with prevalent and incident RA-ILD were assessed using multivariable regression models adjusting for established RA-ILD risk factors. RESULTS Among 2,739 participants with RA (88% male, mean age of 64 years), there were 114 with prevalent and 136 with incident RA-ILD (average time to diagnosis: 6.6 years). Higher IgM anti-MAA-collagen (per 1 SD: adjusted odds ratio [aOR] 1.28, 95% confidence interval [CI] 1.02-1.61), IgA anti-MAA-fibrinogen (aOR 1.48, 95% CI 1.14-1.92), and IgA (aOR 1.78, 95% CI 1.34-2.37) and IgG (aOR 1.48, 95% CI 1.14-1.92) anti-MAA-vimentin antibodies were associated with prevalent RA-ILD. In incident analyses, higher IgA (per one SD: adjusted hazards ratio [aHR] 1.40, 95% CI 1.11-1.76) and IgM (aHR 1.29, 95% CI 1.04-1.60) anti-MAA-albumin antibody concentrations were associated with increased ILD risk. Participants with IgA (aHR 2.13, 95% CI 1.16-3.90) or IgM (aHR 1.98, 95% CI 1.08-3.64) anti-MAA-albumin antibody concentrations in the highest quartile had an approximately two-fold increased risk of incident RA-ILD. Across all isotypes, anti-MAA-fibrinogen, anti-MAA-collagen, and anti-MAA-vimentin antibodies were not significantly associated with incident RA-ILD. CONCLUSION Protein-specific anti-MAA antibodies to collagen, fibrinogen, and vimentin were associated with prevalent RA-ILD. IgA and IgM anti-MAA-albumin antibodies were associated with a higher risk of incident RA-ILD. These findings suggest that MAA modifications and resultant immune responses may contribute to RA-ILD pathogenesis.
Collapse
Affiliation(s)
- Nozima Aripova
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
| | - Geoffrey M. Thiele
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Michael J. Duryee
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Carlos D. Hunter
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Yangyuna Yang
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Punyasha Roul
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Dana P. Ascherman
- Division of Rheumatology, Pittsburgh VA and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Scott M. Matson
- Division of Pulmonary, Critical Care and Sleep, University of Kansas, Kansas City, KS, USA
| | - Gary Kunkel
- Division of Rheumatology, University of Utah and VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Grant W. Cannon
- Division of Rheumatology, University of Utah and VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Katherine D. Wysham
- Division of Rheumatology, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| | - Gail S. Kerr
- Division of Rheumatology, Washington D.C. Veterans Affairs Medical Center, Georgetown and Howard University Hospitals, Washington, DC, USA
| | | | - Joshua F. Baker
- Division of Rheumatology, Corporal Michael J. Crescenz VA and University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jill A. Poole
- Division of Allergy & Immunology, University of Nebraska Medical Center, Omaha, NE USA
| | - Ted R. Mikuls
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Bryant R. England
- Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
9
|
Xu J, Hu H, Sun Y, Zhao Z, Zhang D, Yang L, Lu Q. The fate of immune complexes in membranous nephropathy. Front Immunol 2024; 15:1441017. [PMID: 39185424 PMCID: PMC11342396 DOI: 10.3389/fimmu.2024.1441017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
The most characteristic feature of membranous nephropathy (MN) is the presence of subepithelial electron dense deposits and the consequential thickening of the glomerular basement membrane. There have been great advances in the understanding of the destiny of immune complexes in MN by the benefit of experimental models represented by Heymann nephritis. Subepithelial immune complexes are formed in situ by autoantibodies targeting native autoantigens or exogenous planted antigens such as the phospholipase A2 receptor (PLA2R) and cationic BSA respectively. The nascent immune complexes would not be pathogenic until they develop into immune deposits. Podocytes are the major source of autoantigens in idiopathic membranous nephropathy. They also participate in the modulation and removal of the immune complexes to a large extent. The balance between deposition and clearance is regulated by a wide range of factors such as the composition and physicochemical properties of the immune complexes and the complement system. Complement components such as C3 and C1q have been reported to be precipitated with the deposits whereas a complement regulatory protein CR1 expressed by podocytes is involved in the phagocytosis of immune complexes by podocytes. Podocytes regulate the dynamic change of immune complexes which is disturbed in membranous nephropathy. To elucidate the precise fate of the immune complexes is essential for developing more rational and novel therapies for membranous nephropathy.
Collapse
Affiliation(s)
- Jie Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Haikun Hu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhe Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zihan Zhao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Danyuan Zhang
- Qi Huang of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Yang
- Department of Nephropathy, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Qingyi Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Toyoda T, Mankia K. Prevention of Rheumatoid Arthritis in At-Risk Individuals: Current Status and Future Prospects. Drugs 2024; 84:895-907. [PMID: 38954266 DOI: 10.1007/s40265-024-02061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
Early intervention has been the cornerstone of improving outcomes in patients with rheumatoid arthritis. Over the past decade, the boundaries have been pushed in an attempt to achieve effective prevention strategies in those who are at high risk of developing rheumatoid arthritis. Core risk factors including the presence of serum anti-citrullinated protein antibodies, arthralgia and subclinical inflammation on imaging are highly predictive of arthritis development. The influence of air pollution, diet and the role of microbiome on disease progression are less clear. In turn, therapeutic focus has shifted to an earlier pre-arthritis phase of the disease continuum where the clinically apparent arthritis may potentially be intercepted. Seven proof-of-concept interventional trials in at-risk individuals have been conducted so far. Whether true prevention of rheumatoid arthritis is possible remains elusive. Promising signals towards permanent disease modulation and improvement in symptom burden were seen with some immunomodulatory therapies, whilst others were unsuccessful. Long-term follow-up is required to ascertain a true effect. Looking forward, a better understanding of the natural history and underlying biological mechanisms of arthritis development and more accurate, validated risk stratification is needed.
Collapse
Affiliation(s)
- Task Toyoda
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
11
|
El-Gabalawy H. The Impact of Rheumatoid Arthritis on First Nations and How We Can Work With Communities to Prevent It. J Rheumatol 2024; 51:3-9. [PMID: 38950968 DOI: 10.3899/jrheum.2024-0369_dunlop-dottridge] [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] [Accepted: 04/09/2024] [Indexed: 07/03/2024]
Abstract
Rheumatoid arthritis (RA) is prevalent in many Indigenous North American First Nations (FN) and tends to be seropositive, familial, and disabling, as well as associated with highly unfavorable outcomes such as early mortality. The risk of developing RA is based on a perfect storm of gene-environment interactions underpinning this risk. The gene-environment interactions include a high frequency of shared epitope encoding HLA alleles, particularly HLA-DRB1*1402, in the background population, and prevalent predisposing environmental factors such as smoking and periodontal disease. Together, these provide a compelling rationale for an RA prevention agenda in FN communities. Our research team has worked in partnership with several FN communities to prospectively follow the first-degree relatives of FN patients with RA, with the aim of better understanding the preclinical stages of RA in this population. We have focused on specific features of the anticitrullinated protein antibodies (ACPA) and other proteomic biomarkers as predictors of future development of RA. These studies have now led us to consider interventions having a favorable risk-benefit ratio if applied at a stage prior to a hypothetical "point of no return," when the autoimmunity potentially becomes irreversible. Based on a supportive mouse model and available human studies of curcumin, omega-3, and vitamin D supplements, we are undertaking studies where we screen communities using dried blood spot technology adapted for the detection of ACPA, and then enrolling ACPA-positive individuals in studies that use a combination of these supplements. These studies are guided by shared decision-making principles.
Collapse
Affiliation(s)
- Hani El-Gabalawy
- H. El-Gabalawy, MD, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
12
|
Raposo B, Klareskog L, Robinson WH, Malmström V, Grönwall C. The peculiar features, diversity and impact of citrulline-reactive autoantibodies. Nat Rev Rheumatol 2024; 20:399-416. [PMID: 38858604 DOI: 10.1038/s41584-024-01124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
Since entering the stage 25 years ago as a highly specific serological biomarker for rheumatoid arthritis, anti-citrullinated protein antibodies (ACPAs) have been a topic of extensive research. This hallmark B cell response arises years before disease onset, displays interpatient autoantigen variability, and is associated with poor clinical outcomes. Technological and scientific advances have revealed broad clonal diversity and intriguing features including high levels of somatic hypermutation, variable-domain N-linked glycosylation, hapten-like peptide interactions, and clone-specific multireactivity to citrullinated, carbamylated and acetylated epitopes. ACPAs have been found in different isotypes and subclasses, in both circulation and tissue, and are secreted by both plasmablasts and long-lived plasma cells. Notably, although some disease-promoting features have been reported, results now demonstrate that certain monoclonal ACPAs therapeutically block arthritis and inflammation in mouse models. A wealth of functional studies using patient-derived polyclonal and monoclonal antibodies have provided evidence for pathogenic and protective effects of ACPAs in the context of arthritis. To understand the roles of ACPAs, one needs to consider their immunological properties by incorporating different facets such as rheumatoid arthritis B cell biology, environmental triggers and chronic antigen exposure. The emerging picture points to a complex role of citrulline-reactive autoantibodies, in which the diversity and dynamics of antibody clones could determine clinical progression and manifestations.
Collapse
Affiliation(s)
- Bruno Raposo
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Vivianne Malmström
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
13
|
Takada H, Demoruelle MK, Deane KD, Nakamura S, Katsumata Y, Ikari K, Buckner JH, Robinson WH, Seifert JA, Feser ML, Moss L, Norris JM, Harigai M, Hsieh EW, Holers VM, Okamoto Y. Expansion of HLA-DR Positive Peripheral Helper T and Naive B Cells in Anticitrullinated Protein Antibody-Positive Individuals At Risk for Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1023-1035. [PMID: 38412870 PMCID: PMC11213678 DOI: 10.1002/art.42839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/23/2023] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVE To investigate immune dysregulation in the peripheral blood that contributes to the pre-rheumatoid arthritis (RA) stage of RA development in anticitrullinated protein antibody (ACPA)+ individuals. METHODS Using 37 markers by mass cytometry, we investigated peripheral blood mononuclear cells (PBMCs) from ACPA+ at-risk individuals, ACPA+ early untreated patients with RA, and ACPA- controls in the Tokyo Women's Medical University cohort (n = 17 in each group). Computational algorithms, FlowSOM and Optimized t-Distributed Stochastic Neighbor Embedding, were employed to explore specific immunologic differences between study groups. These findings were further evaluated, and longitudinal changes were explored, using flow cytometry and PBMCs from the US-based Targeting Immune Responses for Prevention of RA cohort that included 11 ACPA+ individuals who later developed RA (pre-RA), of which 9 had post-RA diagnosis PBMCs (post-RA), and 11 ACPA- controls. RESULTS HLA-DR+ peripheral helper T (Tph) cells, activated regulatory T cells, PD-1hi CD8+ T cells, and CXCR5-CD11c-CD38+ naive B cells were significantly expanded in PBMCs from at-risk individuals and patients with early RA from the Tokyo Women's Medical University cohort. Expansion of HLA-DR+ Tph cells and CXCR5-CD11c-CD38+ naive B cells was likewise found in both pre-RA and post-RA time points in the Targeting Immune Responses for Prevention of RA cohort. CONCLUSION The expansion of HLA-DR+ Tph cells and CXCR5-CD11c-CD38+ naive B cells in ACPA+ individuals, including those who developed inflammatory arthritis and classified RA, supports a key role of these cells in transition from pre-RA to classified RA. These findings may identify a new mechanistic target for treatment and prevention in RA.
Collapse
Affiliation(s)
- Hideto Takada
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M. Kristen Demoruelle
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin D. Deane
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Shohei Nakamura
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Yasuhiro Katsumata
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopedic Surgery, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | | | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
| | - Jennifer A. Seifert
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Marie L. Feser
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - LauraKay Moss
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Masayoshi Harigai
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| | - Elena W.Y. Hsieh
- University of Colorado School of Medicine, Department of Immunology and Microbiology Aurora, CO, USA
- University of Colorado School of Medicine, Children’s Hospital Colorado, Department of Pediatrics, Section of Allergy & Immunology, Aurora, CO, USA
| | - V. Michael Holers
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yuko Okamoto
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women’s Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Taylor PC, Schett G, Huizinga TW, Wang Q, Ibrahim F, Zhou B, Liva SG, Shaik JSB, Xiong Y, Leu JH, Panchakshari RA, Loza MJ, Ma K, Dhatt H, Rojo Cella R, Karyekar CS, Cuff CA, Gao S, Fei K. Nipocalimab, an anti-FcRn monoclonal antibody, in participants with moderate to severe active rheumatoid arthritis and inadequate response or intolerance to anti-TNF therapy: results from the phase 2a IRIS-RA study. RMD Open 2024; 10:e004278. [PMID: 38942592 PMCID: PMC11227837 DOI: 10.1136/rmdopen-2024-004278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
OBJECTIVES To investigate the efficacy, safety, pharmacokinetics and pharmacodynamics of nipocalimab in participants with moderate to severe active rheumatoid arthritis (RA) and inadequate response or intolerance to ≥1 antitumour necrosis factor agent. METHODS In this phase 2a study, participants with RA seropositive for anticitrullinated protein antibodies (ACPA) or rheumatoid factors were randomised 3:2 to nipocalimab (15 mg/kg intravenously every 2 weeks) or placebo from Weeks 0 to 10. Efficacy endpoints (primary endpoint: change from baseline in Disease Activity Score 28 using C reactive protein (DAS28-CRP) at Week 12) and patient-reported outcomes (PROs) were assessed through Week 12. Safety, pharmacokinetics and pharmacodynamics were assessed through Week 18. RESULTS 53 participants were enrolled (nipocalimab/placebo, n=33/20). Although the primary endpoint did not reach statistical significance for nipocalimab versus placebo, a numerically higher change from baseline in DAS28-CRP at Week 12 was observed (least squares mean (95% CI): -1.03 (-1.66 to -0.40) vs -0.58 (-1.24 to 0.07)), with numerically higher improvements in all secondary efficacy outcomes and PROs. Serious adverse events were reported in three participants (burn infection, infusion-related reaction and deep vein thrombosis). Nipocalimab significantly and reversibly reduced serum immunoglobulin G, ACPA and circulating immune complex levels but not serum inflammatory markers, including CRP. ACPA reduction was associated with DAS28-CRP remission and 50% response rate in American College of Rheumatology (ACR) criteria; participants with a higher baseline ACPA had greater clinical improvement. CONCLUSIONS Despite not achieving statistical significance in the primary endpoint, nipocalimab showed consistent, numerical efficacy benefits in participants with moderate to severe active RA, with greater benefit observed for participants with a higher baseline ACPA. TRIAL REGISTRATION NUMBER NCT04991753.
Collapse
Affiliation(s)
- Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Georg Schett
- Department of Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU), Nürnberg, Germany
| | | | - Qingmin Wang
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | | | - Bei Zhou
- Janssen Research & Development, LLC, Chesterbrook, Pennsylvania, USA
| | - Sophia G Liva
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | | | - Yuan Xiong
- Janssen Research & Development, LLC, Raritan, New Jersey, USA
| | - Jocelyn H Leu
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | | | - Matthew J Loza
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Keying Ma
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Harman Dhatt
- Janssen Pharmaceutical Companies of Johnson & Johnson, Scottsdale, Arizona, USA
| | | | - Chetan S Karyekar
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Carolyn A Cuff
- Janssen Research & Development, LLC, Cambridge, Massachusetts, USA
| | - Sheng Gao
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Kaiyin Fei
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
15
|
Bongers KS, Massett A, O'Dwyer DN. The Oral-Lung Microbiome Axis in Connective Tissue Disease-Related Interstitial Lung Disease. Semin Respir Crit Care Med 2024; 45:449-458. [PMID: 38626906 DOI: 10.1055/s-0044-1785673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Connective tissue disease-related interstitial lung disease (CTD-ILD) is a frequent and serious complication of CTD, leading to high morbidity and mortality. Unfortunately, its pathogenesis remains poorly understood; however, one intriguing contributing factor may be the microbiome of the mouth and lungs. The oral microbiome, which is a major source of the lung microbiome through recurrent microaspiration, is altered in ILD patients. Moreover, in recent years, several lines of evidence suggest that changes in the oral and lung microbiota modulate the pulmonary immune response and thus may play a role in the pathogenesis of ILDs, including CTD-ILD. Here, we review the existing data demonstrating oral and lung microbiota dysbiosis and possible contributions to the development of CTD-ILD in rheumatoid arthritis, Sjögren's syndrome, systemic sclerosis, and systemic lupus erythematosus. We identify several areas of opportunity for future investigations into the role of the oral and lung microbiota in CTD-ILD.
Collapse
Affiliation(s)
- Kale S Bongers
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Angeline Massett
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - David N O'Dwyer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Sherman MA, Farhadi PN, Pak K, Trieu EP, Sarkar K, Targoff IN, Neely ML, Mammen AL, Rider LG. Myositis-Associated Autoantibodies in Patients With Juvenile Myositis Are Associated With Refractory Disease and Mortality. Arthritis Rheumatol 2024; 76:963-972. [PMID: 38272842 PMCID: PMC11136598 DOI: 10.1002/art.42813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
OBJECTIVE Myositis-associated autoantibodies (MAAs) have been associated with overlap myositis, certain disease manifestations such as interstitial lung disease (ILD), and worse prognosis in the idiopathic inflammatory myopathies. MAAs overall remain largely uncharacterized in patients with juvenile-onset myositis. Moreover, it is unknown whether the number of MAAs is associated with disease severity. METHODS Patients with juvenile myositis in cross-sectional natural history studies who underwent testing for myositis autoantibodies were included. Demographics, myositis autoantibodies, clinical characteristics, medications received, and outcomes of those with and without MAAs were compared. Multivariable logistic regression was performed to determine whether the number of MAAs detected was associated with severe disease features. RESULTS Among 551 patients, 36% had an MAA and 13% had more than one MAA. Among those who were MAA positive, there was a higher frequency of overlap myositis (18% vs 5.9%, P < 0.001). MAA positivity was associated with certain clinical features, including Raynaud phenomenon (odds ratio [OR] 2.44, 95% confidence interval [CI] 1.41-4.28) and ILD (OR 3.43, 95% CI 1.75-6.96), as well as a chronic disease course (OR 1.72, 95% CI 1.10-2.72) and mortality (OR 3.76, 95% CI 1.72-8.43). The number of MAAs was also associated with mortality (OR 1.83, 95% CI 1.16-2.86). CONCLUSION MAAs were prevalent in a large cohort of patients with juvenile myositis. ILD, refractory disease, and mortality were associated with MAA positivity. Prospective studies are needed to determine whether early detection of MAAs may lead to improved outcomes for patients with juvenile myositis.
Collapse
Affiliation(s)
- Matthew A. Sherman
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Payam Noroozi Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Pak
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Edward P. Trieu
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kakali Sarkar
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Ira N. Targoff
- Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, and Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Megan L. Neely
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Andrew L. Mammen
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa G. Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
17
|
Leavy OC, Kawano-Dourado L, Stewart ID, Quint JK, Solomon JJ, Borie R, Crestani B, Wain LV, Jenkins G, Dieudé P, Minelli C. Rheumatoid arthritis and idiopathic pulmonary fibrosis: a bidirectional Mendelian randomisation study. Thorax 2024; 79:538-544. [PMID: 38649271 PMCID: PMC11137470 DOI: 10.1136/thorax-2023-220856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/07/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND A usual interstitial pneumonia (UIP) pattern of lung injury is a key feature of idiopathic pulmonary fibrosis (IPF) and is also observed in up to 40% of individuals with rheumatoid arthritis (RA)-associated interstitial lung disease (RA-ILD). The RA-UIP phenotype could result from either a causal relationship of RA on UIP or vice versa, or from a simple co-occurrence of RA and IPF due to shared demographic, genetic or environmental risk factors. METHODS We used two-sample bidirectional Mendelian randomisation (MR) to test the hypothesis of a causal effect of RA on UIP and of UIP on RA, using variants from genome-wide association studies (GWAS) of RA (separately for seropositive (18 019 cases and 991 604 controls) and seronegative (8515 cases and 1 015 471 controls) RA) and of IPF (4125 cases and 20 464 controls) as genetic instruments. Sensitivity analyses were conducted to assess the robustness of the results to violations of the MR assumptions. FINDINGS IPF showed a significant causal effect on seropositive RA, with developing IPF increasing the risk of seropositive RA (OR=1.06, 95% CI: 1.04 to 1.08, p<0.001) which was robust under all models. For the MR in the other direction, seropositive RA showed a significant protective effect on IPF (OR=0.93; 95% CI: 0.87 to 0.99; p=0.032), but the effect was not significant when sensitivity analyses were applied. This was likely because of bias due to exclusion of patients with RA from among the cases in the IPF GWAS, or possibly because our genetic instruments did not fully capture the effect of the complex human leucocyte antigen region, the strongest RA genetic risk factor. INTERPRETATION Our findings support the hypothesis that RA-UIP may be due to a cause-effect relationship between UIP and RA, rather than due to a coincidental occurrence of IPF in patients with RA. The significant causal effect of IPF on seropositive RA suggests that pathomechanisms involved in the development of UIP may promote RA, and this may help inform future guidelines on screening for ILD in patients with RA.
Collapse
Affiliation(s)
- Olivia C Leavy
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Leticia Kawano-Dourado
- Hcor Research Institute, Hcor, São Paulo, Brazil
- Pulmonary Division, Heart Institute (InCor), University of Sao Paulo, Sao Paulo, Brazil
| | - Iain D Stewart
- National Heart and Lung Institute, Imperial College London, London, UK
- National Institue of Health and Care Research, Imperial Biomedical Research Unit, Imperial College London, London, UK
| | - Jennifer K Quint
- National Heart and Lung Institute, Imperial College London, London, UK
- National Institutue of Health and Care Excellence Imperial Biomedical Research Unit, Imperial College London, London, UK
| | - Joshua J Solomon
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Raphael Borie
- Service de Pneumologie A Hôpital Bichat, APHP, Paris, France
- Université Paris Cité, Inserm, PHERE, Paris, France
| | - Bruno Crestani
- Service de Pneumologie A Hôpital Bichat, APHP, Paris, France
- Université Paris Cité, Inserm, PHERE, Paris, France
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, UK
- National Institue of Health and Care Research, Imperial Biomedical Research Unit, Imperial College London, London, UK
| | - Philippe Dieudé
- Service de Pneumologie A Hôpital Bichat, APHP, Paris, France
- Université Paris Cité, Inserm, PHERE, Paris, France
| | - Cosetta Minelli
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
18
|
Thomas MA, Naik P, Wang H, Giles JT, Girgis AA, Kim SY, Johnson TP, Curran AM, Crawford JD, Jahanbani S, Bingham CO, Robinson WH, Na CH, Darrah E. The monocyte cell surface is a unique site of autoantigen generation in rheumatoid arthritis. Proc Natl Acad Sci U S A 2024; 121:e2304199121. [PMID: 38630712 PMCID: PMC11047081 DOI: 10.1073/pnas.2304199121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Although anti-citrullinated protein autoantibodies (ACPAs) are a hallmark serological feature of rheumatoid arthritis (RA), the mechanisms and cellular sources behind the generation of the RA citrullinome remain incompletely defined. Peptidylarginine deiminase IV (PAD4), one of the key enzymatic drivers of citrullination in the RA joint, is expressed by granulocytes and monocytes; however, the subcellular localization and contribution of monocyte-derived PAD4 to the generation of citrullinated autoantigens remain underexplored. In this study, we demonstrate that PAD4 displays a widespread cellular distribution in monocytes, including expression on the cell surface. Surface PAD4 was enzymatically active and capable of citrullinating extracellular fibrinogen and endogenous surface proteins in a calcium dose-dependent manner. Fibrinogen citrullinated by monocyte-surface PAD4 could be specifically recognized over native fibrinogen by a panel of eight human monoclonal ACPAs. Several unique PAD4 substrates were identified on the monocyte surface via mass spectrometry, with citrullination of the CD11b and CD18 components of the Mac-1 integrin complex being the most abundant. Citrullinated Mac-1 was found to be a target of ACPAs in 25% of RA patients, and Mac-1 ACPAs were significantly associated with HLA-DRB1 shared epitope alleles, higher C-reactive protein and IL-6 levels, and more erosive joint damage. Our findings implicate the monocyte cell surface as a unique and consequential site of extracellular and cell surface autoantigen generation in RA.
Collapse
Affiliation(s)
- Mekha A. Thomas
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Pooja Naik
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Hong Wang
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Jon T. Giles
- Division of Rheumatology, Columbia University, College of Physicians and Surgeons, New York, NY10032
| | - Alexander A. Girgis
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21224
| | - Seok-Young Kim
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Tory P. Johnson
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD20892
| | - Ashley M. Curran
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Jonathan D. Crawford
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA94304
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA94550
| | - Clifton O. Bingham
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA94304
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA94550
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Erika Darrah
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| |
Collapse
|
19
|
Di Matteo A, Mankia K, Garcia-Montoya L, Sharrack S, Duquenne L, Nam JL, Mahler M, Emery P. Utility of testing for third-generation anticyclic citrullinated peptide (anti-CCP3) antibodies in individuals who present with new musculoskeletal symptoms but have a negative second-generation anticyclic citrullinated peptide (anti-CCP2) antibody test. RMD Open 2024; 10:e003927. [PMID: 38599655 PMCID: PMC11015229 DOI: 10.1136/rmdopen-2023-003927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/22/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVES To investigate the role of third-generation anticyclic citrullinated peptide (anti-CCP3) antibodies in predicting progression to inflammatory arthritis (IA) in individuals with new musculoskeletal (MSK) symptoms and a negative second-generation anti-CCP antibody test (anti-CCP2-). METHODS 469 anti-CCP2- individuals underwent baseline anti-CCP3 testing (QUANTA Lite CCP3; Inova Diagnostics) and received a post enrolment 12-month questionnaire. A rheumatologist confirmed or excluded diagnosis of IA. Univariable/multivariable analyses were performed to assess the value of anti-CCP3 in predicting IA development in these anti-CCP2- individuals. RESULTS Only 16/469 (3.4%) anti-CCP2- individuals had a positive anti-CCP3 test. Of these 16 individuals, 4 developed IA. In addition, 61/469 (13.0%) anti-CCP2- individuals self-reported, to have developed, IA. Progression was confirmed in 43/61 of them (70.5%); of whom 30/43 (69.8%) and 13/43 (30.2%) were given a diagnosis of IA and rheumatoid arthritis (RA), respectively. In qualitative univariable analysis, anti-CCP3 positivity was associated with self-reported progression (p<0.01) and IA (p=0.03), but not with RA. Anti-CCP3 levels differed significantly between progressors and non-progressors (p<0.01) for all three categories. At the manufacturer's cut-off, OR for progression ranged from 2.4 (95% CI 0.5 to 18.6; RA) to 7.5 (95% CI 2.3 to 24.0; self-reported progression). Interestingly, when cut-offs for anti-CCP3 were optimised, lower values (≥5 units) significantly increased the OR for progression in all three categories. In multivariable analysis, anti-CCP3 positivity at the manufacturer's cut-off did not remain associated with IA progression, while this lower cut-off value (≥5 units) was associated with diagnosis of RA (p=0.02). CONCLUSIONS Anti-CCP3 testing could improve the prediction of IA development in anti-CCP2- individuals with new MSK symptoms.
Collapse
Affiliation(s)
- Andrea Di Matteo
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Leticia Garcia-Montoya
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sana Sharrack
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Laurence Duquenne
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | - Michael Mahler
- Werfen Autoimmunity Technology Center, San Diego, California, USA
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
20
|
van Dijk BT, Boeren AMP, Khidir SJH, den Hollander NK, van der Helm-van Mil AHM. Work-related physical strain and development of joint inflammation in the trajectory of emerging inflammatory and rheumatoid arthritis: a prospective cohort study. RMD Open 2024; 10:e003895. [PMID: 38599648 PMCID: PMC11015171 DOI: 10.1136/rmdopen-2023-003895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/27/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVES Rheumatoid arthritis (RA) mainly affects small joints. Despite the mechanical function of joints, the role of mechanical stress in the development of arthritis is insufficiently understood. We hypothesised that mechanical stress/physical strain is a risk factor for joint inflammation in RA. Therefore, we studied work-related physical strain in subjects with clinically suspected arthralgia (CSA) as a risk factor for the presence of imaging-detected subclinical joint inflammation and the development of clinical arthritis/RA. METHODS In 501 CSA patients and 155 symptom-free persons' occupation-related physical strain was quantified using the International Standard Classification of Occupations. Contrast-enhanced hand-MRIs were made and evaluated for joint inflammation (sum of synovitis/tenosynovitis/osteitis). CSA patients were followed on RA development. Age relationship was studied using an interaction term of physical strain with age. RESULTS The degree of physical strain in CSA is associated with the severity of joint inflammation, independent of educational-level/BMI/smoking (interaction physical strain-age p=0.007; indicating a stronger association with increasing age). Physical strain is associated with higher tenosynovitis scores, in particular. In symptom-free persons, physical strain was not associated with imaging-detected joint inflammation. Higher degrees of physical strain also associated with higher risks for RA development in an age-dependent manner (HR=1.20 (1.06-1.37)/10-year increase in age), independent of educational-level/BMI/smoking. This association was partly mediated by an effect via subclinical joint inflammation. CONCLUSIONS Work-related physical strain increases the risk of subclinical joint inflammation and of developing RA. The age relationship suggests an effect of long-term stress or that tenosynovium is more sensitive to stress at older age. Together, the data indicate that mechanical stress contributes to the development of arthritis in RA.
Collapse
Affiliation(s)
| | - Anna M P Boeren
- Rheumatology, LUMC, Leiden, The Netherlands
- Rheumatology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
21
|
Jiao M, Zhang Y, Song X, Xu B. The role and mechanism of TXNDC5 in disease progression. Front Immunol 2024; 15:1354952. [PMID: 38629066 PMCID: PMC11019510 DOI: 10.3389/fimmu.2024.1354952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Thioredoxin domain containing protein-5 (TXNDC5), also known as endothelial protein-disulfide isomerase (Endo-PDI), is confined to the endoplasmic reticulum through the structural endoplasmic reticulum retention signal (KDEL), is a member of the PDI protein family and is highly expressed in the hypoxic state. TXNDC5 can regulate the rate of disulfide bond formation, isomerization and degradation of target proteins through its function as a protein disulfide isomerase (PDI), thereby altering protein conformation, activity and improving protein stability. Several studies have shown that there is a significant correlation between TXNDC5 gene polymorphisms and genetic susceptibility to inflammatory diseases such as rheumatoid, fibrosis and tumors. In this paper, we detail the expression characteristics of TXNDC5 in a variety of diseases, summarize the mechanisms by which TXNDC5 promotes malignant disease progression, and summarize potential therapeutic strategies to target TXNDC5 for disease treatment.
Collapse
Affiliation(s)
- Mingxia Jiao
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Province Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yeyong Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong, China
| | - Xie Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bing Xu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Province Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
22
|
Aripova N, Kremer JM, Pappas DA, Reed G, England BR, Robinson BH, Curtis JR, Thiele GM, Mikuls TR. Anti-citrullinated protein antibody profiles predict changes in disease activity in patients with rheumatoid arthritis initiating biologics. Rheumatology (Oxford) 2024; 63:542-550. [PMID: 37252826 PMCID: PMC10836988 DOI: 10.1093/rheumatology/kead260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/01/2023] [Accepted: 05/17/2023] [Indexed: 06/01/2023] Open
Abstract
OBJECTIVES To determine whether an expanded antigen-specific ACPA profile predicts changes in disease activity in patients with RA initiating biologics. METHODS The study included participants from a prospective, non-randomized, observational RA cohort. For this sub-study, treatment groups of interest included biologic-naïve initiating anti-TNF, biologic-exposed initiating non-TNF, and biologic-naïve initiating abatacept. ACPAs to 25 citrullinated peptides were measured using banked enrolment serum. Principal component analysis (PCA) was performed and associations of resulting principal component (PC) scores (in quartiles) and anti-CCP3 antibody (≤15, 16-250 or >250 U/ml) with EULAR (good/moderate/none) treatment response at 6 months were examined using adjusted ordinal regression models. RESULTS Participants (n = 1092) had a mean age of 57 (13) years and 79% were women. At 6 months, 68.5% achieved a moderate/good EULAR response. There were three PCs that cumulatively explained 70% of variation in ACPA values. In models including the three components and anti-CCP3 antibody category, only PC1 and PC2 were associated with treatment response. The highest quartile for PC1 (odds ratio [OR] 1.76; 95% CI: 1.22, 2.53) and for PC2 (OR 1.74; 95% CI: 1.23, 2.46) were associated with treatment response after multivariable adjustment. There was no evidence of interaction between PCs and treatment group in EULAR responses (P-value for interaction >0.1). CONCLUSION An expanded ACPA profile appears to be more strongly associated with biologic treatment response in RA than commercially available anti-CCP3 antibody levels. However, further enhancements to PCA will be needed to effectively prioritize between different biologics available for the treatment of RA.
Collapse
Affiliation(s)
- Nozima Aripova
- Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joel M Kremer
- CorEvitas LLC, Waltham, MA, USA
- The Corrona Research Foundation, Albany, NY, USA
- Department of Medicine, Center for Rheumatology, Albany Medical College, Albany, NY, USA
| | - Dimitrios A Pappas
- CorEvitas LLC, Waltham, MA, USA
- The Corrona Research Foundation, Albany, NY, USA
- Division of Rheumatology, Columbia University, New York, NY, USA
| | - George Reed
- CorEvitas LLC, Waltham, MA, USA
- The Corrona Research Foundation, Albany, NY, USA
- Department of Medicine, University of Massachusetts, Worcester, MA, USA
| | - Bryant R England
- Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Bill H Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine & VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Jeffrey R Curtis
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Geoffrey M Thiele
- Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Ted R Mikuls
- Division of Rheumatology, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
23
|
Gomez AM, Brewer RC, Moon JS, Acharya S, Kongpachith S, Wang Q, Jahanbani S, Wong HH, Lanz TV, Love ZZ, Min-Oo G, Niedziela-Majka A, Robinson WH. Anti-Citrullinated Protein Antibodies With Multiple Specificities Ameliorate Collagen Antibody-Induced Arthritis in a Time-Dependent Manner. Arthritis Rheumatol 2024; 76:181-191. [PMID: 37610274 DOI: 10.1002/art.42679] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/23/2023] [Accepted: 08/17/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVE Anti-citrullinated protein antibodies (ACPAs) are highly specific for rheumatoid arthritis (RA) and have long been regarded as pathogenic. Despite substantial in vitro evidence supporting this claim, reports investigating the proinflammatory effects of ACPAs in animal models of arthritis are rare and include mixed results. Here, we sequenced the plasmablast antibody repertoire of a patient with RA and functionally characterized the encoded ACPAs. METHODS We expressed ACPAs from the antibody repertoire of a patient with RA and characterized their autoantigen specificities on antigen arrays and enzyme-linked immunosorbent assays. Binding affinities were estimated by bio-layer interferometry. Select ACPAs (n = 9) were tested in the collagen antibody-induced arthritis (CAIA) mouse model to evaluate their effects on joint inflammation. RESULTS Recombinant ACPAs bound preferentially and with high affinity (nanomolar range) to citrullinated (cit) autoantigens (primarily histones and fibrinogen) and to auto-cit peptidylarginine deiminase 4 (PAD4). ACPAs were grouped for in vivo testing based on their predominant cit-antigen specificities. Unexpectedly, injections of recombinant ACPAs significantly reduced paw thickness and arthritis severity in CAIA mice as compared with isotype-matched control antibodies (P ≤ 0.001). Bone erosion, synovitis, and cartilage damage were also significantly reduced (P ≤ 0.01). This amelioration of CAIA was observed for all the ACPAs tested and was independent of cit-PAD4 and cit-fibrinogen specificities. Furthermore, disease amelioration was more prominent when ACPAs were injected at earlier stages of CAIA than at later phases of the model. CONCLUSION Recombinant patient-derived ACPAs ameliorated CAIA. Their antiinflammatory effects were more preventive than therapeutic. This study highlights a potential protective role for ACPAs in arthritis.
Collapse
Affiliation(s)
- Alejandro M Gomez
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - R Camille Brewer
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Jae-Seung Moon
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Suman Acharya
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Sarah Kongpachith
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Qian Wang
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Shaghayegh Jahanbani
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Heidi H Wong
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Tobias V Lanz
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | - Zelda Z Love
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| | | | | | - William H Robinson
- Stanford University School of Medicine, Stanford, and VA Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
24
|
Wang Q, Feng D, Jia S, Lu Q, Zhao M. B-Cell Receptor Repertoire: Recent Advances in Autoimmune Diseases. Clin Rev Allergy Immunol 2024; 66:76-98. [PMID: 38459209 DOI: 10.1007/s12016-024-08984-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Abstract
In the field of contemporary medicine, autoimmune diseases (AIDs) are a prevalent and debilitating group of illnesses. However, they present extensive and profound challenges in terms of etiology, pathogenesis, and treatment. A major reason for this is the elusive pathophysiological mechanisms driving disease onset. Increasing evidence suggests the indispensable role of B cells in the pathogenesis of autoimmune diseases. Interestingly, B-cell receptor (BCR) repertoires in autoimmune diseases display a distinct skewing that can provide insights into disease pathogenesis. Over the past few years, advances in high-throughput sequencing have provided powerful tools for analyzing B-cell repertoire to understand the mechanisms during the period of B-cell immune response. In this paper, we have provided an overview of the mechanisms and analytical methods for generating BCR repertoire diversity and summarize the latest research progress on BCR repertoire in autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), primary Sjögren's syndrome (pSS), multiple sclerosis (MS), and type 1 diabetes (T1D). Overall, B-cell repertoire analysis is a potent tool to understand the involvement of B cells in autoimmune diseases, facilitating the creation of innovative therapeutic strategies targeting specific B-cell clones or subsets.
Collapse
Affiliation(s)
- Qian Wang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Delong Feng
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
25
|
Heutz JW, Rogier C, Niemantsverdriet E, van den Eeden SJF, de Jong PHP, Lubberts E, Geluk A, van der Helm-van Mil AHM. The course of cytokine and chemokine gene expression in clinically suspect arthralgia patients during progression to inflammatory arthritis. Rheumatology (Oxford) 2024; 63:563-570. [PMID: 37280058 PMCID: PMC10836970 DOI: 10.1093/rheumatology/kead238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/14/2023] [Accepted: 05/13/2023] [Indexed: 06/08/2023] Open
Abstract
OBJECTIVES Autoantibody responses increase years before the onset of inflammatory arthritis (IA) and are stable during transitioning from clinically suspect arthralgia (CSA) to IA. Cytokine and chemokine levels also increase years before IA onset. However, the course in the at-risk stage of CSA during progression to disease or non-progression is unknown. To increase the understanding of processes mediating disease development, we studied the course of cytokine, chemokine and related receptors gene expression in CSA patients during progression to IA and in CSA patients who ultimately did not develop IA. METHODS Whole-blood RNA expression of 37 inflammatory cytokines, chemokines and related receptors was determined by dual-colour reverse transcription multiplex ligation-dependent probe amplification in paired samples of CSA patients at CSA onset and either at IA development or after 24 months without IA development. ACPA-positive and ACPA-negative CSA patients developing IA were compared at CSA onset and during progression to IA. Generalised estimating equations tested changes over time. A false discovery rate approach was applied. RESULTS None of the cytokine/chemokine genes significantly changed in expression between CSA onset and IA development. In CSA patients without IA development, G-CSF expression decreased (P = 0.001), whereas CCR6 and TNIP1 expression increased (P < 0.001 and P = 0.002, respectively) over a 2 year period. Expression levels in ACPA-positive and ACPA-negative CSA patients who developed IA were similar. CONCLUSION Whole-blood gene expression of assessed cytokines, chemokines and related receptors did not change significantly from CSA to IA development. This suggests that changes in expression of these molecules may not be related to the final process of developing chronicity and may have occurred preceding CSA onset. Changes in gene expression in CSA patients without IA development may provide clues for processes related to resolution.
Collapse
Affiliation(s)
- Judith W Heutz
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Cleo Rogier
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Susan J F van den Eeden
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Pascal H P de Jong
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Erik Lubberts
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Annette H M van der Helm-van Mil
- Department of Rheumatology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
26
|
Rudbaek JJ, Agrawal M, Torres J, Mehandru S, Colombel JF, Jess T. Deciphering the different phases of preclinical inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2024; 21:86-100. [PMID: 37950021 PMCID: PMC11148654 DOI: 10.1038/s41575-023-00854-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/12/2023]
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory disease (IMID) of the gastrointestinal tract and includes two subtypes: Crohn's disease and ulcerative colitis. It is well-recognized that IBD is associated with a complex multifactorial aetiology that includes genetic predisposition and environmental exposures, with downstream dysregulation of systemic immune function and host-microbial interactions in the local environment in the gut. Evidence to support the notion of a multistage development of IBD is growing, as has been observed in other IMIDs such as rheumatoid arthritis and systemic lupus erythematosus. With the rising worldwide incidence of IBD, it is increasingly important to understand the complex interplay of pathological events during the different stages of disease development to enable IBD prediction and prevention strategies. In this article, we review comprehensively the current evidence pertaining to the preclinical phase of IBD, including at-risk, initiation and expansion phases. We also discuss the framework of preclinical IBD, expanding on underlying pathways in IBD development, future research directions and IBD development in the context of other IMIDs.
Collapse
Affiliation(s)
- Jonas J Rudbaek
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Section for Biomarkers, Immunology and Antibodies, Department for Congenital Disorders, Statens Serum Institut, Copenhangen, Denmark
| | - Manasi Agrawal
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joana Torres
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
- Division of Gastroenterology, Hospital da Luz, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Saurabh Mehandru
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tine Jess
- Center for Molecular Prediction of Inflammatory Bowel Disease, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark.
- Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
27
|
Sherif AE, Sajid-ur-Rehman M, Asif M, Qadeer I, Khan KUR. Anti-inflammatory, analgesic, and antipyretic potential of Oxystelma esculentum (L. f.) Sm. using in vitro, in vivo, and in silico studies. Front Pharmacol 2024; 14:1326968. [PMID: 38293669 PMCID: PMC10825028 DOI: 10.3389/fphar.2023.1326968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
The objective of the current study was to evaluate the anti-inflammatory, analgesic, and antipyretic potential of Oxystelma esculentum using different animal models. The phytochemical profile was determined by assessing its total phenolic content (TPC) and total flavonoid content (TFC), followed by the high-performance liquid chromatography (HPLC) technique. The in vitro anti-inflammatory potential of O. esculentum ethanolic extract (OEE) was evaluated by lipoxygenase enzyme inhibition activity and a human red blood cell (HRBC) membrane stability assay. The in vivo anti-inflammatory potential of the plant was determined by the carrageenan-induced paw edema test, and the analgesic potential by the hot plate test, tail-flick test, formalin-induced analgesia, acetic acid-induced writhing activities, and yeast-induced elevation of body temperature. The values of total phenolic content (212.6 ± 3.18 µg GAE/g) and total flavonoid content (37.6 ± 1.76 µg QE/g) were observed. The results showed that OEE exhibited significant antioxidant capacity in DPPH (2,2-diphenyl-1-picrylhydrazyl) (266.3 ± 7.35 μmol TE/g), ABTS (2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (1,066.3 ± 7.53 μmol TE/g), and FRAP (ferric reducing antioxidant power) (483.6 ± 3.84 μmol TE/g) assays. The HPLC analysis demonstrated phytocompounds with anti-inflammatory potential, such as chlorogenic acid, gallic acid, 4-hydroxybenzoic acid, caffeic acid, ferulic acid, and coumarin. The plant showed in vitro anti-inflammatory activity through the inhibition of lipoxygenase enzyme with a high percentage (56.66%) and HRBC membrane stability (67.29%). In in vivo studies, OEE exhibited significant (p < 0.05) anti-inflammatory (carrageenan-induced paw edema model), analgesic (hot plate test, tail-flick test, formalin-induced analgesia, and acetic acid-induced writhing), and antipyretic (rectal temperature reduction) responses at different doses (100, 300, and 500 mg/kg). Molecular docking studies showed significant binding affinities of phytocompounds compared to indomethacin and predicted various binding interactions for stable conformations. The results of in vitro, in vivo, and in silico studies supported the anti-inflammatory, analgesic, and antipyretic potential of O. esculentum.
Collapse
Affiliation(s)
- Asmaa E. Sherif
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Muhammad Sajid-ur-Rehman
- Department of Pharmacognosy, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Asif
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Iram Qadeer
- Department of Zoology, Government Sadiq College Women University, Bahawalpur, Pakistan
| | - Kashif ur Rehman Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
28
|
van Steenbergen HW, Cope AP, van der Helm-van Mil AHM. Rheumatoid arthritis prevention in arthralgia: fantasy or reality? Nat Rev Rheumatol 2023; 19:767-777. [PMID: 37814057 DOI: 10.1038/s41584-023-01035-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2023] [Indexed: 10/11/2023]
Abstract
The concept of a 'window of opportunity' in treating a disease assumes the existence of a time frame during which the trajectory of the disease can be effectively and permanently modified. In rheumatoid arthritis (RA), optimal timing of this period is presumed to be during the phase before arthritis is clinically apparent and disease is diagnosed. Several proof-of-concept trials of treatment during the 'arthralgia' phase of RA have been completed in the past 4 years, with the underlying notion that temporary treatment at this stage could prevent the development of RA or induce a sustained reduction in the burden of disease. This Review summarizes the results of these trials and reflects on the outcomes in relation to the patients' perspectives. Overall, the majority of symptomatic at-risk individuals could benefit from a fixed period treatment, even if RA does not develop. Various factors must be taken into consideration when translating these findings into clinical practice. More evidence is needed to target the individuals at highest risk, and additional tools are needed to monitor treatment and guide decisions about whether treatment can be discontinued. Without these tools, there is a paradoxical risk of seemingly increasing the incidence of the disease and prolonging disease duration, which is the opposite of what the concept of intervening in the window of opportunity entails.
Collapse
Affiliation(s)
| | - Andrew P Cope
- Centre for Rheumatic Diseases, Department of Inflammation Biology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Annette H M van der Helm-van Mil
- Department of Rheumatology, Leiden University Medical Centre, Leiden, the Netherlands.
- Department of Rheumatology, Erasmus Medical Centre, Rotterdam, the Netherlands.
| |
Collapse
|
29
|
James EA, Holers VM, Iyer R, Prideaux EB, Rao NL, Rims C, Muir VS, Posso SE, Bloom MS, Zia A, Elliott SE, Adamska JZ, Ai R, Brewer RC, Seifert JA, Moss L, Barzideh S, Demoruelle MK, Striebich CC, Okamoto Y, Sainbayar E, Crook AA, Peterson RA, Vanderlinden LA, Wang W, Boyle DL, Robinson WH, Buckner JH, Firestein GS, Deane KD. Multifaceted immune dysregulation characterizes individuals at-risk for rheumatoid arthritis. Nat Commun 2023; 14:7637. [PMID: 37993439 PMCID: PMC10665556 DOI: 10.1038/s41467-023-43091-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
Molecular markers of autoimmunity, such as antibodies to citrullinated protein antigens (ACPA), are detectable prior to inflammatory arthritis (IA) in rheumatoid arthritis (RA) and may define a state that is 'at-risk' for future RA. Here we present a cross-sectional comparative analysis among three groups that include ACPA positive individuals without IA (At-Risk), ACPA negative individuals and individuals with early, ACPA positive clinical RA (Early RA). Differential methylation analysis among the groups identifies non-specific dysregulation in peripheral B, memory and naïve T cells in At-Risk participants, with more specific immunological pathway abnormalities in Early RA. Tetramer studies show increased abundance of T cells recognizing citrullinated (cit) epitopes in At-Risk participants, including expansion of T cells reactive to citrullinated cartilage intermediate layer protein I (cit-CILP); these T cells have Th1, Th17, and T stem cell memory-like phenotypes. Antibody-antigen array analyses show that antibodies targeting cit-clusterin, cit-fibrinogen and cit-histone H4 are elevated in At-Risk and Early RA participants, with the highest levels of antibodies detected in those with Early RA. These findings indicate that an ACPA positive at-risk state is associated with multifaceted immune dysregulation that may represent a potential opportunity for targeted intervention.
Collapse
Affiliation(s)
- Eddie A James
- Benaroya Research Institute, Seattle, WA, 98101, USA
| | - V Michael Holers
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Radhika Iyer
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | - E Barton Prideaux
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Navin L Rao
- Janssen Research and Development, Spring House, PA, 19477, USA
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA, 98101, USA
| | | | | | - Michelle S Bloom
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | - Amin Zia
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | - Serra E Elliott
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | - Julia Z Adamska
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | - Rizi Ai
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - R Camille Brewer
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | - Jennifer A Seifert
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - LauraKay Moss
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Saman Barzideh
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - M Kristen Demoruelle
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Christopher C Striebich
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yuko Okamoto
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Division of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Enkhtsogt Sainbayar
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Alexandra A Crook
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ryan A Peterson
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Lauren A Vanderlinden
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - David L Boyle
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94304, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94550, USA
| | | | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
30
|
Hughes-Austin JM, Katz R, Majka DS, Criqui MH, Robinson WH, Firestein GS, Hundley WG, Ix JH. Serum reactivity to citrullinated protein/peptide antigens and left ventricular structure and function in the Multi-Ethnic Study of Atherosclerosis (MESA). PLoS One 2023; 18:e0291967. [PMID: 37874814 PMCID: PMC10597499 DOI: 10.1371/journal.pone.0291967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 09/10/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Antibodies to citrullinated protein antigens have been linked to altered left ventricular (LV) structure and function in patients with rheumatoid arthritis (RA). Serum reactivity to several citrullinated protein/peptide antigens has been identified in RA, which are detectable years before RA onset and in individuals who may never develop RA. Among community-living individuals without heart failure (HF) at baseline in the Multi-Ethnic Study of Atherosclerosis (MESA), we investigated associations between serum reactivity to citrullinated protein/peptide antigens, LV mass, LV ejection fraction (LVEF), and incident HF. METHODS Among 1232 MESA participants, we measured serum reactivity to 28 different citrullinated proteins/peptides using a multiplex bead-based array. Each antibody was defined as having extremely high reactivity (EHR) if >95th percentile cut-off in MESA. Number of EHR antibody responses to citrullinated protein/peptide antigens were summed for each participant (range 0-28). LV mass(g) and LVEF(%) were measured on cardiac MRI. Associations between EHR antibodies and LV mass and LVEF were evaluated using linear regression. Cox proportional hazards models were used to evaluate associations between EHR antibodies and incident HF during 11 years of follow-up, adjusting for age, gender, race/ethnicity, smoking status, systolic blood pressure, use of anti-hypertensive medications, self-reported arthritis, IL-6, body surface area, and estimated glomerular filtration rate. RESULTS Mean age was 65±10, 50% were female, 40% were White, 21% were Black, 26% were Hispanic/Latino, and 14% were Chinese. Twenty-seven percent of MESA participants had extremely high reactivity to ≥ 1 citrullinated protein/peptide antigen. In fully adjusted analysis, every additional EHR antibody was significantly associated with 0.1% lower LVEF (95% CI: -0.17%, -0.02%). No association was observed with LV mass (β per additional EHR antibody) = 0.13±0.15 (p = 0.37)). Neither the presence nor number of EHR antibodies was associated with incident HF during follow-up (HR per additional EHR antibody = 1.008 (95% CI: 0.97, 1.05)). CONCLUSION Greater number of extremely highly reactive antibodies was associated with lower LVEF, but not with LV mass or incident HF. Thus, serum reactivity to citrullinated protein/peptide antigens was associated with subtle subclinical changes in myocardial contractility, but the significance in relation to clinically apparent HF is uncertain.
Collapse
Affiliation(s)
- Jan M. Hughes-Austin
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California, United States of America
| | - Ronit Katz
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Darcy S. Majka
- Division of Rheumatology, DuPage Medical Group, Chicago, Illinois, United States of America
| | - Michael H. Criqui
- Department of Family Medicine and Public Health, University of California, San Diego, La Jolla, California, United States of America
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Gary S. Firestein
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - W. Gregory Hundley
- Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
31
|
England BR. The Multimorbidity Web in rheumatoid arthritis. Rheumatology (Oxford) 2023; 62:SI242-SI251. [PMID: 37871922 DOI: 10.1093/rheumatology/kead246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/17/2023] [Indexed: 10/25/2023] Open
Abstract
Multimorbidity, the presence of multiple chronic conditions, is highly prevalent in people with RA. An essential characteristic of multimorbidity is the interrelatedness of the different conditions that may develop in a multimorbid person. Recent studies have begun to identify and describe the Multimorbidity Web by elucidating unique multimorbidity patterns in people with RA. The primary multimorbidity patterns in this web are cardiopulmonary, cardiometabolic, and mental health and chronic pain multimorbidity. Once caught in the Multimorbidity Web, the consequences can be devastating, with reduced quality of life, physical function, survival, and treatment responses observed in multimorbid RA persons. The development of effective management and preventive approaches for multimorbidity in people with RA is in its infancy. Determining how best to assess, intervene, and prevent multimorbidity in RA is crucial to optimize long-term outcomes in people with RA.
Collapse
Affiliation(s)
- Bryant R England
- Division of Rheumatology & Immunology, Department of Internal Medicine, VA Nebraska-Western Iowa Health Care System, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
32
|
Page A, Delles M, Nègre D, Costa C, Fusil F, Cosset FL. Engineering B cells with customized therapeutic responses using a synthetic circuit. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:1-14. [PMID: 37359346 PMCID: PMC10285500 DOI: 10.1016/j.omtn.2023.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
The expansion of genetic engineering has brought a new dimension for synthetic immunology. Immune cells are perfect candidates because of their ability to patrol the body, interact with many cell types, proliferate upon activation, and differentiate in memory cells. This study aimed at implementing a new synthetic circuit in B cells, allowing the expression of therapeutic molecules in a temporally and spatially restricted manner that is induced by the presence of specific antigens. This should enhance endogenous B cell functions in terms of recognition and effector properties. We developed a synthetic circuit encoding a sensor (a membrane-anchored B cell receptor targeting a model antigen), a transducer (a minimal promoter induced by the activated sensor), and effector molecules. We isolated a 734-bp-long fragment of the NR4A1 promoter, specifically activated by the sensor signaling cascade in a fully reversible manner. We demonstrate full antigen-specific circuit activation as its recognition by the sensor induced the activation of the NR4A1 promoter and the expression of the effector. Overall, such novel synthetic circuits offer huge possibilities for the treatment of many pathologies, as they are completely programmable; thus, the signal-specific sensors and effector molecules can be adapted to each disease.
Collapse
Affiliation(s)
- Audrey Page
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, 69007 Lyon, France
| | - Marie Delles
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, 69007 Lyon, France
| | - Didier Nègre
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, 69007 Lyon, France
| | - Caroline Costa
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, 69007 Lyon, France
| | - Floriane Fusil
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, 69007 Lyon, France
| | - François-Loïc Cosset
- CIRI - Centre International de Recherche en Infectiologie, University Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 Allée d’Italie, 69007 Lyon, France
| |
Collapse
|
33
|
Rodríguez-Carrio J, Alperi-López M, López P, Pérez-Álvarez ÁI, Robinson GA, Alonso-Castro S, Amigo-Grau N, Atzeni F, Suárez A. Humoral responses against HDL are linked to lipoprotein traits, atherosclerosis, inflammation and pathogenic pathways during early arthritis stages. Rheumatology (Oxford) 2023; 62:2898-2907. [PMID: 36617161 DOI: 10.1093/rheumatology/kead009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/20/2022] [Accepted: 01/02/2023] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Chronic inflammation and immune dysregulation are crucial mechanisms for atherosclerosis in RA. Recent evidence suggests a link via humoral responses against high-density lipoproteins (HDL). This study aimed to characterize the specificity, clinical relevance and emergence of humoral responses against HDL along disease course, especially during the earliest phases of arthritis. METHODS IgG and IgM serum levels of antibodies against HDL (anti-HDL) and apolipoprotein A1 (anti-ApoA1) were measured in 82 early RA patients, 14 arthralgia individuals and 96 controls. Established RA patients (n = 42) were included for validation. Atherosclerosis and vascular stiffness were measured by Doppler ultrasound. Lipoprotein content, particle numbers and size were measured by H-NMR. Cytokines were measured by immunoassays. A cardiometabolic-related protein panel was evaluated using high-throughput targeted proteomics. RESULTS Anti-HDL and anti-ApoA1 responses were increased in early RA compared with controls (both P < 0.001) and were comparable to established disease. Only anti-ApoA1 antibodies were increased in arthralgia. IgG anti-HDL and anti-ApoA1 were associated with unfavourable lipoprotein traits in RA and arthralgia, respectively. A similar picture was observed for inflammatory mediators. No associations with clinical features or risk factors were found. IgG anti-HDL were independently associated with atherosclerosis occurrence in early RA, and outperformed patient stratification over conventional algorithms (mSCORE) and their anti-ApoA1 counterparts. Anti-HDL antibodies correlated with proteins involved in immune activation, remodelling and lipid metabolism pathways in early RA. CONCLUSION Humoral responses against HDL particles are an early event along the arthritis course, although quantitative and qualitative differences can be noticed among stages. These differences informed distinct capacities as biomarkers and underlying pathogenic circuits.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Mercedes Alperi-López
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Patricia López
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | | | - George A Robinson
- Centre for Adolescent Rheumatology Versus Arthritis, Department of Medicine, University College London, London, UK
| | - Sara Alonso-Castro
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Núria Amigo-Grau
- Biosfer Teslab, Reus, Spain
- Department of Basic Medical Sciences, Pere Virgili Health Research Institute (IISPV), Universitat Rovira i Virgili (URV), Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBER-DEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Area of Metabolism, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
34
|
Van der Helm-van Mil AHM. Preventive interventions in individuals at risk for Rheumatoid Arthritis: State of the art and perspectives. Joint Bone Spine 2023; 90:105543. [PMID: 36796580 PMCID: PMC7615889 DOI: 10.1016/j.jbspin.2023.105543] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/17/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023]
Abstract
During the last decade, the outlook for patients with rheumatoid arthritis (RA) has improved greatly, especially for patients with autoantibody-positive RA. To further improve long-term disease outcomes, the field has turned to investigating the efficacy of treatment initiated in the pre-arthritic phase of RA, based on the adage "the sooner the better." In this review, the concept of prevention is evaluated and different risk stages are being examined for their pre-test risks of RA development. These risks affect the post-test risk of biomarkers used at these stages and, consequently, the accuracy with which the risk of RA can be estimated. Furthermore, through their effect on accurate risk stratification, these pre-test risks ultimately also associate with the likelihood of false-negative trial results (the so-called "clinicostatistical tragedy"). Outcome measures to assess preventive effects are evaluated and relate to either the occurrence of disease itself or to the severity of risk factors for RA development. Results of recently completed prevention studies are discussed in the light of these theoretical considerations. The results vary, but clear prevention of RA has not been demonstrated. While some treatments (e.g. methotrexate) persistently reduced symptom severity, physical disability, and the severity of imaging joint inflammation, other treatments were not reported to produce long-lasting effects (hydroxychloroquine, rituximab, atorvastatin). The review concludes with future perspectives regarding the design of new prevention studies and considerations and requirements before findings can be implemented in daily practice in individuals at risk of RA attending rheumatology practices.
Collapse
|
35
|
Ghosh N, Reid P, Aude CA, Kirschman J, Goodman S, Bykerk VP, Lakhanpal A, Rajesh D, Chan KK, Robinson WH, Bass AR. Anticitrullinated peptide antibody epitope expansion and the HLA DRB1 'shared epitope' are less common in seropositive checkpoint inhibitor-induced inflammatory arthritis than in longstanding rheumatoid arthritis. RMD Open 2023; 9:e003012. [PMID: 37355249 PMCID: PMC10314674 DOI: 10.1136/rmdopen-2023-003012] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/22/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) can potentially cause ICI-inflammatory arthritis (ICI-IA), which often resembles rheumatoid arthritis (RA). In this study, we examined the degree of anticitrullinated peptide antibodies (ACPA) epitope expansion in CCP+ICI-IA and patients with RA. METHODS We used clinical data and serum from ICI-IA and patients with RA with early disease as well as longstanding disease. A custom, bead-based antigen array was used to identify IgG ACPA reactivities to 18 putative RA-associated citrullinated proteins. Hierarchical clustering software was used to create a heatmap to identify ACPA levels. Additionally, HLA DRB1 typing was performed on ICI-IA patients as well as controls of patients treated with ICI that did not develop ICI-IA (ICI controls). RESULTS Compared to patients with CCP+RA, patients with CCP+ICI-IA were older (p<0.001), less likely to have positive rheumatoid factor (p<0.001) and had a shorter duration of symptoms (p<0.001). There were less ACPA levels and a lower number of distinct ACPA epitopes in the serum of patients with ICI-IA compared with longstanding patients with RA (p<0.001). Among those tested for HLA DRB1, there were no differences in the frequency of the shared epitope between those with ICI-IA and ICI controls. CONCLUSION Patients with ICI-IA had lower ACPA titres and targeted fewer ACPA epitopes than longstanding patients with RA, and there were no significant differences in the presence of the shared epitope between those that developed ICI-IA and ICI controls. It remains to be determined if ICI-IA represents an accelerated model of RA pathogenesis with ICI triggering a transition from preclinical to clinical disease.
Collapse
Affiliation(s)
- Nilasha Ghosh
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Pankti Reid
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, Illinois, USA
| | - Carlos Andres Aude
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Jessica Kirschman
- Department of Medicine, Stanford University, Stanford, California, USA
| | - Susan Goodman
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Vivian P Bykerk
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Amit Lakhanpal
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Diviya Rajesh
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Karmela K Chan
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | | | - Anne R Bass
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
36
|
Sharma A, Goel A. Pathogenesis of rheumatoid arthritis and its treatment with anti-inflammatory natural products. Mol Biol Rep 2023; 50:4687-4706. [PMID: 37022525 DOI: 10.1007/s11033-023-08406-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023]
Abstract
Introduction Rheumatoid arthritis (RA) is a common autoimmune disease across the globe that is chronic and systemic as well. The disease is linked with autoantibodies and is inflammatory, eventually targeting several molecules along with certain modified self-epitopes. The disease majorly affects the joints of an individual. Rheumatoid arthritis is manifested clinically by polyarthritis linked with the dysfunction of the joints. This chiefly affects the synovial joint lining and is linked with progressive dysfunction, premature death, along with socioeconomic implications. The macrophage activation, along with the activation of certain defense cells, results in a response to self-epitopes that helps in providing a better understanding of the disease pathogenesis. Material and methodology For this review article, papers have been retrieved and reviewed from database including PubMed, Scopus and Web of science. Relevant papers were taken fulfilling the criteria for writing this review article. Results This has resulted in the establishment of several new therapeutic techniques that serve as potential inhibitors of such cells. Researchers have gained an interest in understanding this disease to provide strategies for treatment in the last two decades. This also includes recognition followed by the treatment of the disease at its early stages. Various allopathic treatment approaches often have chronic and toxic teratogenic effects. However, to avoid this issue of toxicity followed by side effects, certain medicinal plants have been used in treating RA. Conclusion Medicinal plants possess active phytoconstituents that entail antioxidants as well as anti-inflammatory properties, making them a helpful alternative to allopathic drugs that are often linked with highly toxic effects. This review paper entails a thorough discussion of the epidemiology, pathophysiology, diagnosis, and management of RA. The paper will also focus on the use of herbal plants in the treatment of the disease to avoid the side effects that generally occur in allopathic treatment.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Uttar Pradesh, 281406, Mathura, India
| | - Anjana Goel
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Uttar Pradesh, 281406, Mathura, India.
| |
Collapse
|
37
|
Vinicki JP, Zamora JLV, Salinas RG, Alarcón GS. How are rheumatologists from Argentina managing patients with arthralgias suspicious for progressing to rheumatoid arthritis? Clin Rheumatol 2023; 42:1479-1483. [PMID: 36977950 DOI: 10.1007/s10067-023-06581-0] [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: 01/05/2023] [Revised: 02/26/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023]
Abstract
Patients with arthralgias who could be at risk of progressing to rheumatoid arthritis (RA) represent a clinical challenge. Recommendations for their management and treatment are lacking. The purpose of the present study was to determine how Argentinean rheumatologists deal with these patients. We developed an anonymous ad hoc survey which was sent to 522 Argentinean rheumatologists. The RA study group of our Argentinean Rheumatology National Society assisted in forwarding the surveys to its members via the internet (e-mail or WhatsApp). The findings of the collected data are presented as descriptive statistics. The questionnaires were completed by 255 rheumatologists (overall response rate of 48.9%), and 97.6% confirmed that their practices had received medical consultations to rule out RA in patients with arthralgias. Ultrasound (US) was the method of first choice (93.7%) as part of the evaluation of these patients. For those in whom US power Doppler signal was present in at least one joint, 93.7% of the participants would start treatment and methotrexate was the first choice (58.1%). In patients with tenosynovitis but no synovitis on US, most rheumatologists would start treatment (89.4%), being NSAIDs the drug of first choice (52.3%). Argentinean rheumatologists evaluate patients with imminent RA and treat them based on their clinical judgment and findings from the US evaluation of affected joints; the drug of first choice for these patients among these rheumatologists was methotrexate. Despite published data of recent clinical trials, recommendations for the management and treatment of these patients are necessary.
Collapse
Affiliation(s)
- Juan Pablo Vinicki
- Sección Reumatología, Hospital de Quilmes, 770, Allison Bell, Quilmes, 1878, Buenos Aires, Argentina.
| | | | | | - Graciela S Alarcón
- Marnix E. Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
38
|
Abstract
Various scores have attempted to predict the onset of rheumatoid arthritis (RA). In particular, EULAR proposed a simple rule to identify new-onset arthralgia suspicious for progression to RA. However, its specificity would likely be higher if serological tests were included. In patients with clinical arthritis, reliable predictive criteria for progression to RA have also been identified. Overall, the validity of the available scores is still being debated. Such scores do not fully account for the interactions between risk factors in specific subpopulations. New technologies could help to overcome these limitations, but we need databases containing a sufficient number of RA and pre-RA patients, including pre-diagnostic monitoring. Today, the existing predictive rules cannot compete with expert opinions.
Collapse
|
39
|
Brewer RC, Lanz TV, Hale CR, Sepich-Poore GD, Martino C, Swafford AD, Carroll TS, Kongpachith S, Blum LK, Elliott SE, Blachere NE, Parveen S, Fak J, Yao V, Troyanskaya O, Frank MO, Bloom MS, Jahanbani S, Gomez AM, Iyer R, Ramadoss NS, Sharpe O, Chandrasekaran S, Kelmenson LB, Wang Q, Wong H, Torres HL, Wiesen M, Graves DT, Deane KD, Holers VM, Knight R, Darnell RB, Robinson WH, Orange DE. Oral mucosal breaks trigger anti-citrullinated bacterial and human protein antibody responses in rheumatoid arthritis. Sci Transl Med 2023; 15:eabq8476. [PMID: 36812347 PMCID: PMC10496947 DOI: 10.1126/scitranslmed.abq8476] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 02/02/2023] [Indexed: 02/24/2023]
Abstract
Periodontal disease is more common in individuals with rheumatoid arthritis (RA) who have detectable anti-citrullinated protein antibodies (ACPAs), implicating oral mucosal inflammation in RA pathogenesis. Here, we performed paired analysis of human and bacterial transcriptomics in longitudinal blood samples from RA patients. We found that patients with RA and periodontal disease experienced repeated oral bacteremias associated with transcriptional signatures of ISG15+HLADRhi and CD48highS100A2pos monocytes, recently identified in inflamed RA synovia and blood of those with RA flares. The oral bacteria observed transiently in blood were broadly citrullinated in the mouth, and their in situ citrullinated epitopes were targeted by extensively somatically hypermutated ACPAs encoded by RA blood plasmablasts. Together, these results suggest that (i) periodontal disease results in repeated breaches of the oral mucosa that release citrullinated oral bacteria into circulation, which (ii) activate inflammatory monocyte subsets that are observed in inflamed RA synovia and blood of RA patients with flares and (iii) activate ACPA B cells, thereby promoting affinity maturation and epitope spreading to citrullinated human antigens.
Collapse
Affiliation(s)
- R. Camille Brewer
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Tobias V. Lanz
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Caryn R. Hale
- Rockefeller University, New York City, NY 10065, USA
| | | | - Cameron Martino
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Austin D. Swafford
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Thomas S. Carroll
- Bioinformatics Resource Center, Rockefeller University, 1230 York Ave., New York, NY 10065, USA
| | - Sarah Kongpachith
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Lisa K. Blum
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Serra E. Elliott
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Nathalie E. Blachere
- Rockefeller University, New York City, NY 10065, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | | | - John Fak
- Rockefeller University, New York City, NY 10065, USA
| | - Vicky Yao
- Department of Computer Science, Rice University, Houston, TX 77005, USA
- Department of Computer Science, Princeton University, Princeton, NJ, 08544, USA
| | - Olga Troyanskaya
- Department of Computer Science, Princeton University, Princeton, NJ, 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
- Flatiron Institute, Simons Foundation, New York, NY, 10010, USA
| | - Mayu O. Frank
- Rockefeller University, New York City, NY 10065, USA
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Alejandro M. Gomez
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Radhika Iyer
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Nitya S. Ramadoss
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Orr Sharpe
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | - Lindsay B. Kelmenson
- Division of Rheumatology, University of Colorado - Denver, Aurora, CO, 80045, USA
| | - Qian Wang
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Heidi Wong
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | | | - Mark Wiesen
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Dana T. Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kevin D. Deane
- Division of Rheumatology, University of Colorado - Denver, Aurora, CO, 80045, USA
| | - V. Michael Holers
- Division of Rheumatology, University of Colorado - Denver, Aurora, CO, 80045, USA
| | - Rob Knight
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Robert B. Darnell
- Rockefeller University, New York City, NY 10065, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Dana E. Orange
- Rockefeller University, New York City, NY 10065, USA
- Hospital for Special Surgery, New York City, NY 10075, USA
| |
Collapse
|
40
|
Rheumatoid arthritis and non-coding RNAs; how to trigger inflammation. Life Sci 2023; 315:121367. [PMID: 36639050 DOI: 10.1016/j.lfs.2023.121367] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/31/2022] [Accepted: 12/31/2022] [Indexed: 01/12/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic and chronic inflammatory disease categorized by continuous synovitis in the joints and systemic inflammatory responses that can cause lifelong disability. The major cause of RA is the dysregulation of the immune response. The development of RA disease includes multiplex association of several interleukins and cells, which leads to synovial cell growth, cartilage and bone damage. The primary stage of RA disease is related to the modification of both the innate and adaptive immune systems, which leads to the formation of autoantibodies. This process results in many damaged molecules and epitope spreading. Both the innate (e.g., dendritic cells, macrophages, and neutrophils) and acquired immune cells (e.g., T and B lymphocytes) will increase and continue the chronic inflammatory condition in the next stages of the RA disease. In recent years, non-coding RNAs have been proved as significant controllers of biological functions, especially immune cell expansion and reactions. Non-coding RNAs were primarily containing microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA). Various studies confirmed non-coding RNAs as hopeful markers for diagnosing and curing RA. This review will describe and cover existing knowledge about RA pathogenesis, which might be favorable for discovering possible ncRNA markers for RA.
Collapse
|
41
|
Affiliation(s)
- Ellen M Gravallese
- From the Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston (E.M.G.); and the Division of Rheumatology, Allergy, and Immunology, University of California at San Diego School of Medicine, La Jolla (G.S.F.)
| | - Gary S Firestein
- From the Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston (E.M.G.); and the Division of Rheumatology, Allergy, and Immunology, University of California at San Diego School of Medicine, La Jolla (G.S.F.)
| |
Collapse
|
42
|
Raj R, Thomas S, Gorantla V. Accelerated atherosclerosis in rheumatoid arthritis: a systematic review. F1000Res 2023; 11:466. [PMID: 36249997 PMCID: PMC9551388 DOI: 10.12688/f1000research.112921.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Background: Rheumatoid arthritis (RA) is a highly prevalent, chronic inflammatory condition of the synovial joints that affects approximately 1% of the global population. The pathogenesis of RA is predominantly inflammatory in nature, thereby accelerating the co-occurrence of other immunoinflammatory conditions such as atherosclerosis. Apart from traditional cardiovascular risk factors, RA patients possess a multitude of other factors that predispose them to early atherosclerotic disease. The aim of this systematic review is to assess the prevalence of premature atherosclerosis in RA patients and elucidate the role that proinflammatory cytokines, RA-related autoantibodies, and endothelial dysfunction play in the pathophysiology of RA-mediated atherosclerosis. We also discussed novel biomarkers that can be used to predict early atherosclerosis in RA and current guidelines used to treat RA. Methods: This review followed the PRISMA guidelines to select and analyze relevant articles. A literature search for articles was performed on February 25, 2022, through three research databases including PubMed, ProQuest, and ScienceDirect. The query used to identify relevant publications was "Rheumatoid arthritis and atherosclerosis" and the search duration was set from 2012-2022. Relevant articles were selected based on the inclusion and exclusion criteria. Results: Our initial search generated 21,235 articles. We narrowed our search according to the inclusion and exclusion criteria. After assessing eligibility based on the full content of the articles, 73 articles were ultimately chosen for this review. Conclusion: There is an increased prevalence of accelerated atherosclerosis among RA patients. We found evidence to explain the role of proinflammatory cytokines, RA-related autoantibodies, and endothelial dysfunction in the pathophysiology RA-mediated atherosclerosis. Therapies targeting either the inflammatory load or traditional CV risk-factors seem to improve vascular outcomes in RA patients. Novel markers of atherosclerosis in RA may be useful in predicting premature atherosclerosis and serve as new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Rhea Raj
- St. George's University School of medicine, True Blue, St. George's, Grenada
| | - Sneha Thomas
- University of Maryland Medical Center MTC, Midtown, Baltimore, USA,
| | - Vasavi Gorantla
- St. George's University School of medicine, True Blue, St. George's, Grenada
| |
Collapse
|
43
|
Abstract
Antibody-mediated neurological diseases constitute an emerging clinical entity that remains to be fully explored. Recent studies identified autoantibodies that directly confer pathogenicity, and it was shown that in these cases immunotherapies can result in profound positive patient responses. These advances highlight the urgent need for improved means to effectively screen patient samples for novel autoantibodies (aAbs) and their subsequent characterization. Here, we discuss challenges and opportunities for peptide microarrays to contribute to the identification, mapping, and characterization of the underlying monospecific disease-defining binding surfaces. We outline control experiments, workflow modifications and bioinformatic filtering methods that enhance the predictive power of array-based studies. Further, we highlight experimental and computer-based display approaches that have the potential to expand the use of synthetic microarrays over the detection of discontinuous epitopes. Knowledge over the autoantibody epitopes in neurological disease will enhance our understanding of the pathological mechanisms and thereby potentially contribute to novel diagnostic approaches or even innovative antigen-specific treatments that avoid the serious adverse effects seen with currently used immunosuppressive therapies.
Collapse
Affiliation(s)
- Ivan Talucci
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
44
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM, Kuhn KA. Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022; 14. [PMID: 36288282 PMCID: PMC9804515 DOI: 10.1126/scitranslmed.abn5166 10.1126/scitranslmed.abn5166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The mucosal origins hypothesis of rheumatoid arthritis (RA) proposes a central role for mucosal immune responses in the initiation or perpetuation of the systemic autoimmunity that occurs with disease. However, the connection between the mucosa and systemic autoimmunity in RA remains unclear. Using dual immunoglobulin A (IgA) and IgG family plasmablast-derived monoclonal autoantibodies obtained from peripheral blood of individuals at risk for RA, we identified cross-reactivity between RA-relevant autoantigens and bacterial taxa in the closely related families Lachnospiraceae and Ruminococcaceae. After generating bacterial isolates within the Lachnospiraceae/Ruminococcaceae genus Subdoligranulum from the feces of an individual, we confirmed monoclonal antibody binding and CD4+ T cell activation in individuals with RA compared to control individuals. In addition, when Subdoligranulum isolate 7 but not isolate 1 colonized germ-free mice, it stimulated TH17 cell expansion, serum RA-relevant IgG autoantibodies, and joint swelling reminiscent of early RA, with histopathology characterized by antibody deposition and complement activation. Systemic immune responses were likely due to mucosal invasion along with the generation of colon-isolated lymphoid follicles driving increased fecal and serum IgA by isolate 7, because B and CD4+ T cell depletion not only halted intestinal immune responses but also eliminated detectable clinical disease. In aggregate, these findings demonstrate a mechanism of RA pathogenesis through which a specific intestinal strain of bacteria can drive systemic autoantibody generation and joint-centered antibody deposition and immune activation.
Collapse
Affiliation(s)
- Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael R. Clay
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alex Ren Hsu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jennifer Seifert
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marie L. Feser
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA 98101
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth A. Bemis
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | - Daniel N. Frank
- Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - M. Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | | | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,Corresponding Author:
| |
Collapse
|
45
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM, Kuhn KA. Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022; 14:eabn5166. [PMID: 36288282 PMCID: PMC9804515 DOI: 10.1126/scitranslmed.abn5166] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The mucosal origins hypothesis of rheumatoid arthritis (RA) proposes a central role for mucosal immune responses in the initiation or perpetuation of the systemic autoimmunity that occurs with disease. However, the connection between the mucosa and systemic autoimmunity in RA remains unclear. Using dual immunoglobulin A (IgA) and IgG family plasmablast-derived monoclonal autoantibodies obtained from peripheral blood of individuals at risk for RA, we identified cross-reactivity between RA-relevant autoantigens and bacterial taxa in the closely related families Lachnospiraceae and Ruminococcaceae. After generating bacterial isolates within the Lachnospiraceae/Ruminococcaceae genus Subdoligranulum from the feces of an individual, we confirmed monoclonal antibody binding and CD4+ T cell activation in individuals with RA compared to control individuals. In addition, when Subdoligranulum isolate 7 but not isolate 1 colonized germ-free mice, it stimulated TH17 cell expansion, serum RA-relevant IgG autoantibodies, and joint swelling reminiscent of early RA, with histopathology characterized by antibody deposition and complement activation. Systemic immune responses were likely due to mucosal invasion along with the generation of colon-isolated lymphoid follicles driving increased fecal and serum IgA by isolate 7, because B and CD4+ T cell depletion not only halted intestinal immune responses but also eliminated detectable clinical disease. In aggregate, these findings demonstrate a mechanism of RA pathogenesis through which a specific intestinal strain of bacteria can drive systemic autoantibody generation and joint-centered antibody deposition and immune activation.
Collapse
Affiliation(s)
- Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael R. Clay
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alex Ren Hsu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jennifer Seifert
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marie L. Feser
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA 98101
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth A. Bemis
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | - Daniel N. Frank
- Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - M. Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | | | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,Corresponding Author:
| |
Collapse
|
46
|
Serény-Litvai T, Bajnok A, Temesfoi V, Nörenberg J, Pham-Dobor G, Kaposi A, Varnagy A, Kovacs K, Pentek S, Koszegi T, Mezosi E, Berki T. B cells from anti-thyroid antibody positive, infertile women show hyper-reactivity to BCR stimulation. Front Immunol 2022; 13:1039166. [DOI: 10.3389/fimmu.2022.1039166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Anti-thyroid antibody (ATA) positivity affects 1 out of 9 women in childbearing age and presents a significant risk for infertility. Emerging evidence indicates that alterations in the B cell receptor induced calcium (Ca2+) signaling could be key in the development of autoimmunity. We aimed to investigate the Ca2+ flux response of B lymphocyte subsets to BCR stimulation in Hashimoto’s thyroiditis and related infertility. We collected peripheral blood samples from ATA+, infertile, euthyroid patients (HIE), hypothyroid, ATA+ patients before (H1) and after levothyroxine treatment (H2), and age-matched healthy controls (HC). All B cell subsets of ATA+, infertile, euthyroid patients showed elevated basal Ca2+ level and hyper-responsivity to BCR ligation compared to the other groups, which could reflect altered systemic immune function. The Ca2+ flux of hypothyroid patients was similar to healthy controls. The levothyroxine-treated patients had decreased prevalence of CD25+ B cells and lower basal Ca2+ level compared to pre-treatment. Our results support the role of altered Ca2+ flux of B cells in the early phase of thyroid autoimmunity and infertility.
Collapse
|
47
|
Dei Zotti F, Moriconi C, Qiu A, Miller A, Hudson KE. Distinct CD4+ T cell signature in ANA-positive young adult patients. Front Immunol 2022; 13:972127. [PMCID: PMC9608560 DOI: 10.3389/fimmu.2022.972127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
Failure of immune tolerance can lead to autoantibody production resulting in autoimmune diseases, a broad spectrum of organ-specific or systemic disorders. Immune tolerance mechanisms regulate autoreactive T and B cells, yet some lymphocytes escape and promote autoantibody production. CD4+ T cell dysregulation, characterized by decreased or impaired regulatory cells (Tregs) and/or accumulation of memory and effector T cells such as TH17, plays a crucial role in the pathogenesis of these diseases. Antinuclear antibody (ANAs) testing is used as a first step for the diagnosis of autoimmune disorders, although most ANA-positive individuals do not have nor will develop an autoimmune disease. Studying the differences of T cell compartment among healthy blood donors, ANA-negative patients and ANA-positive patients, in which loss of tolerance have not led to autoimmunity, may improve our understanding on how tolerance mechanisms fail. Herein, we report that ANA-positive patients exhibit a distinct distribution of T cell subsets: significantly reduced frequencies of recent thymic emigrants (RTE) and naïve T cells, and significantly increased frequencies of central memory T cells, TH2 and TH17 cells; modulations within the T cell compartment are most profound within the 18-40 year age range. Moreover, CD4+ T cells in ANA-positive patients are metabolically active, as determined by a significant increase in mTORC1 and mTORC2 signals, compared to ANA-negative patients and healthy blood donors. No significant impairment of Treg numbers or pro-inflammatory cytokine production was observed. These results identify a unique T cell signature associated with autoantibody production in the absence of autoimmune disease.
Collapse
|
48
|
O’Neil LJ, Meng X, Mcfadyen C, Fritzler MJ, El-Gabalawy HS. Serum proteomic networks associate with pre-clinical rheumatoid arthritis autoantibodies and longitudinal outcomes. Front Immunol 2022; 13:958145. [PMID: 36159862 PMCID: PMC9492875 DOI: 10.3389/fimmu.2022.958145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Objectives The development of autoantibody directed towards citrullinated proteins (ACPA) are predictive of RA in at-risk individuals. The biological events that underpin loss of immune tolerance and progression into inflammatory arthritis are not known. We sought to identify serum proteomic alterations that drive autoantibody formation, persistence and progression into inflammatory arthritis in a cohort of first-degree relatives (FDR) of RA patients. Methods We studied baseline serum samples from a cohort of Indigenous FDR (n = 147) and quantified serum proteins using a 48-plex platform. Longitudinal outcomes were defined on the basis of ACPA status and progression into inflammatory arthritis (IA). K-means clustering, differential expression, and principal components analyze group differences. A co-expression module analysis was used to identify enriched networks. Random forest was used to classify ACPA positive samples, while network analysis was used to understand underlying biological processes based on protein expression. Results We defined 6 proteomic clusters, with enrichment of ACPA positive samples in one of the clusters. 23 of 24 differentially expressed proteins in ACPA positive samples were upregulated. A co-expression network was enriched in ACPA positive sera and individuals who progressed into IA. Random Forest achieved an area under the curve of 0.767 to classify ACPA positive sera in a test dataset. Network analysis revealed upregulation of JAK-STAT signalling as being activated in those at highest risk to develop future IA. Conclusions The serum proteome provides a rich dataset to understand biological processes in ACPA seropositive individuals. A combination of serum biomarkers, including ACPA, may predict future arthritis onset in at-risk individuals.
Collapse
Affiliation(s)
- Liam J. O’Neil
- Department of Internal Medicine, Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
- *Correspondence: Liam J. O’Neil,
| | - Xiaobo Meng
- Department of Internal Medicine, Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
| | - Caitlin Mcfadyen
- Department of Internal Medicine, Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
| | | | - Hani S. El-Gabalawy
- Department of Internal Medicine, Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
49
|
Holers VM, Kuhn KA, Demoruelle MK, Norris JM, Firestein GS, James EA, Robinson WH, Buckner JH, Deane KD. Mechanism-driven strategies for prevention of rheumatoid arthritis. RHEUMATOLOGY & AUTOIMMUNITY 2022; 2:109-119. [PMID: 36312783 PMCID: PMC9610829 DOI: 10.1002/rai2.12043] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023]
Abstract
In seropositive rheumatoid arthritis (RA), the onset of clinically apparent inflammatory arthritis (IA) is typically preceded by a prolonged period of autoimmunity manifest by the presence of circulating autoantibodies that can include antibodies to citrullinated protein antigens (ACPA) and rheumatoid factor (RF). This period prior to clinical IA can be designated preclinical RA in those individuals who have progressed to a clinical diagnosis of RA, and an 'at-risk' status in those who have not developed IA but exhibit predictive biomarkers of future clinical RA. With the goal of developing RA prevention strategies, studies have characterized immune phenotypes of preclinical RA/at-risk states. From these studies, a model has emerged wherein mucosal inflammation and dysbiosis may lead first to local autoantibody production that should normally be transient, but instead is followed by systemic spread of the autoimmunity as manifest by serum autoantibody elevations, and ultimately drives the development of clinically identified joint inflammation. This model can be envisioned as the progression of disease development through serial 'checkpoints' that in principle should constrain or resolve autoimmunity; however, instead the checkpoints 'fail' and clinical RA develops. Herein we review the immune processes that are likely to be present at each step and the potential therapeutic strategies that could be envisioned to delay, diminish, halt or even reverse the progression to clinical RA. Notably, these prevention strategies could utilize existing therapies approved for clinical RA, therapies approved for other diseases that target relevant pathways in the preclinical/at-risk state, or approaches that target novel pathways.
Collapse
Affiliation(s)
- V. Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kristine A. Kuhn
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - M. Kristen Demoruelle
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jill M. Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Gary S. Firestein
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305, USA and VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | | | - Kevin D. Deane
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
50
|
Prognostic alternative splicing events related splicing factors define the tumor microenvironment and pharmacogenomic landscape in lung adenocarcinoma. Aging (Albany NY) 2022; 14:6689-6715. [PMID: 36006412 PMCID: PMC9467413 DOI: 10.18632/aging.204244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 08/09/2022] [Indexed: 12/05/2022]
Abstract
Background: Recent studies identified correlations between splicing factors (SFs) and tumor progression and therapy. However, the potential roles of SFs in immune regulation and the tumor microenvironment (TME) remain unknown. Methods: We used UpSet plots to screen for prognostic-related alternative splicing (AS) events. We evaluated SF patterns in specific immune landscapes. Single sample gene set enrichment analysis (ssGSEA) algorithms were used to quantify relative infiltration levels in immune cell subsets. Principal component analysis (PCA) algorithm-based SFscore were used to evaluate SF patterns in individual tumors with an immune response. Results: From prognosis-related AS events, 16 prognosis-related SFs were selected to construct three SF patterns. Further TME analyses showed these patterns were highly consistent with immune-inflamed, immune-excluded, and immune-desert landscapes. Based on SFscore constructed using differentially expressed genes (DEGs) between SF patterns, patients were classified into two immune-subtypes associated with differential pharmacogenomic landscapes and cell features. A low SFscore was associated with high immune cell infiltration, high tumor mutation burden (TMB), and elevated expression of immune check points (ICPs), indicating a better immune response. Conclusions: SFs are significantly associated with TME remodeling. Evaluating different SF patterns enhances our understanding of the TME and improves effective immunotherapy strategies.
Collapse
|