دورية أكاديمية

Antibodies to Malondialdehyde-Acetaldehyde Adduct Are Associated With Prevalent and Incident Rheumatoid Arthritis-Associated Interstitial Lung Disease in US Veterans.

التفاصيل البيبلوغرافية
العنوان: Antibodies to Malondialdehyde-Acetaldehyde Adduct Are Associated With Prevalent and Incident Rheumatoid Arthritis-Associated Interstitial Lung Disease in US Veterans.
المؤلفون: Aripova N; University of Nebraska Medical Center, Omaha., Thiele GM; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha., Duryee MJ; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha., Hunter CD; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha., Yang Y; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha., Roul P; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha., Ascherman DP; Pittsburgh Veterans Affairs and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania., Matson SM; University of Kansas, Kansas City., Kunkel G; University of Utah and Veterans Affairs Salt Lake City Health Care System, Salt Lake City., Cannon GW; University of Utah and Veterans Affairs Salt Lake City Health Care System, Salt Lake City., Wysham KD; Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle., Kerr GS; Washington D.C. Veterans Affairs Medical Center, Georgetown and Howard University Hospitals., Monach PA; Boston Veterans Affairs Health Care System, Boston, Massachusetts., Baker JF; Corporal Michael J. Crescenz Veterans Affairs and University of Pennsylvania, Perelman School of Medicine, Philadelphia., Poole JA; University of Nebraska Medical Center, Omaha., Mikuls TR; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha., England BR; University of Nebraska Medical Center and Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha.
المصدر: Arthritis & rheumatology (Hoboken, N.J.) [Arthritis Rheumatol] 2024 May 20. Date of Electronic Publication: 2024 May 20.
Publication Model: Ahead of Print
نوع المنشور: Journal Article
اللغة: English
بيانات الدورية: Publisher: Wiley Country of Publication: United States NLM ID: 101623795 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 2326-5205 (Electronic) Linking ISSN: 23265191 NLM ISO Abbreviation: Arthritis Rheumatol Subsets: MEDLINE
أسماء مطبوعة: Original Publication: Malden, MA : Wiley, [2014]-
مستخلص: 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.
(© 2024 The Author(s). Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)
References: Bongartz T, Nannini C, Medina‐Velasquez YF, et al. Incidence and mortality of interstitial lung disease in rheumatoid arthritis: a population‐based study. Arthritis Rheum 2010;62(6):1583–1591.
Olson AL, Swigris JJ, Sprunger DB, et al. Rheumatoid arthritis‐interstitial lung disease‐associated mortality. Am J Respir Crit Care Med 2011;183(3):372–378.
Raimundo K, Solomon JJ, Olson AL, et al. Rheumatoid arthritis‐interstitial lung disease in the United States: prevalence, incidence, and healthcare costs and mortality. J Rheumatol 2019;46(4):360–369.
Hyldgaard C, Hilberg O, Pedersen AB, et al. A population‐based cohort study of rheumatoid arthritis‐associated interstitial lung disease: comorbidity and mortality. Ann Rheum Dis 2017;76(10):1700–1706.
Sparks JA, Chang SC, Liao KP, et al. Rheumatoid arthritis and mortality among women during 36 years of prospective follow‐up: results from the Nurses’ Health Study. Arthritis Care Res (Hoboken) 2016;68(6):753–762.
Johnson TM, Yang Y, Roul P, et al. A narrowing mortality gap: temporal trends of cause‐specific mortality in a national, matched cohort study in U.S. veterans with rheumatoid arthritis. Arthritis Care Res (Hoboken) 2023;75(8):1648–1658.
Van Kalsbeek D, Brooks R, Shaver D, et al. Peripheral blood biomarkers for rheumatoid arthritis‐associated interstitial lung disease: a systematic review. ACR Open Rheumatol 2023;5(4):201–226.
Ingegnoli F, Castelli R, Gualtierotti R. Rheumatoid factors: clinical applications. Dis Markers 2013;35(6):727–734.
Aggarwal R, Liao K, Nair R, Ringold S, Costenbader KH. Anti‐citrullinated peptide antibody assays and their role in the diagnosis of rheumatoid arthritis. Arthritis Rheum 2009;61(11):1472–1483.
Xie S, Li S, Chen B, et al. Serum anti‐citrullinated protein antibodies and rheumatoid factor increase the risk of rheumatoid arthritis‐related interstitial lung disease: a meta‐analysis. Clin Rheumatol 2021;40(11):4533–4543.
Natalini JG, Baker JF, Singh N, et al. Autoantibody seropositivity and risk for interstitial lung disease in a prospective male‐predominant rheumatoid arthritis cohort of U.S. veterans. Ann Am Thorac Soc 2021;18(4):598–605.
Harlow L, Rosas IO, Gochuico BR, et al. Identification of citrullinated Hsp90 isoforms as novel autoantigens in rheumatoid arthritis‐associated interstitial lung disease. Arthritis Rheum 2013;65(4):869–879.
Liu Y, Liu C, Li L, et al. High levels of antibodies to citrullinated α‐enolase peptide‐1 (CEP‐1) identify erosions and interstitial lung disease (ILD) in a Chinese rheumatoid arthritis cohort. Clin Immunol 2019;200:10–15.
Castellanos‐Moreira R, Rodríguez‐García SC, Gomara MJ, et al. Anti‐carbamylated proteins antibody repertoire in rheumatoid arthritis: evidence of a new autoantibody linked to interstitial lung disease. Ann Rheum Dis 2020;79(5):587–594.
England BR, Duryee MJ, Roul P, et al. Malondialdehyde‐acetaldehyde adducts and antibody responses in rheumatoid arthritis‐associated interstitial lung disease. Arthritis Rheumatol 2019;71(9):1483–1493.
Darrah E, Giles JT, Davis RL, et al. Autoantibodies to Peptidylarginine Deiminase 2 are associated with less severe disease in rheumatoid arthritis. Front Immunol 2018;9:2696.
Luedders BA, Wheeler AM, Ascherman DP, et al. Plasma matrix metalloproteinase concentrations and risk of interstitial lung disease in a prospective rheumatoid arthritis cohort. Arthritis Rheumatol Published online January 25, 2024. doi:https://doi.org/10.1002/art.42812.
Juge P‐A, Lee JS, Ebstein E, et al. MUC5B promoter variant and rheumatoid arthritis with interstitial lung disease. N Engl J Med 2018;379(23):2209–2219.
Wheeler AM, Baker JF, Poole JA, et al. Genetic, social, and environmental risk factors in rheumatoid arthritis‐associated interstitial lung disease. Semin Arthritis Rheum 2022;57:152098.
Takizawa Y, Suzuki A, Sawada T, et al. Citrullinated fibrinogen detected as a soluble citrullinated autoantigen in rheumatoid arthritis synovial fluids. Ann Rheum Dis 2006;65(8):1013–1020.
Bang H, Egerer K, Gauliard A, et al. Mutation and citrullination modifies vimentin to a novel autoantigen for rheumatoid arthritis. Arthritis Rheum 2007;56(8):2503–2511.
Yoshida M, Tsuji M, Kurosaka D, et al. Autoimmunity to citrullinated type II collagen in rheumatoid arthritis. Mod Rheumatol 2006;16(5):276–281.
Aripova N, Kremer JM, Pappas DA, et al. Anti‐citrullinated protein antibody profiles predict changes in disease activity in patients with rheumatoid arthritis initiating biologics. Rheumatology (Oxford) 2024;63(2):542–550.
Joshua V, Hensvold AH, Reynisdottir G, et al. Association between number and type of different ACPA fine specificities with lung abnormalities in early, untreated rheumatoid arthritis. RMD Open 2020;6(2):e001278.
Ytterberg AJ, Joshua V, Reynisdottir G, et al. Shared immunological targets in the lungs and joints of patients with rheumatoid arthritis: identification and validation. Ann Rheum Dis 2015;74(9):1772–1777.
Baka Z, Buzás E, Nagy G. Rheumatoid arthritis and smoking: putting the pieces together. Arthritis Res Ther 2009;11(4):238.
Rangel‐Moreno J, Hartson L, Navarro C, et al. Inducible bronchus‐associated lymphoid tissue (iBALT) in patients with pulmonary complications of rheumatoid arthritis. J Clin Invest 2006;116(12):3183–3194.
Reynisdottir G, Karimi R, Joshua V, et al. Structural changes and antibody enrichment in the lungs are early features of anti‐citrullinated protein antibody‐positive rheumatoid arthritis. Arthritis Rheumatol 2014;66(1):31–39.
Mikuls TR, Duryee MJ, Rahman R, et al. Enrichment of malondialdehyde‐acetaldehyde antibody in the rheumatoid arthritis joint. Rheumatology (Oxford) 2017;56(10):1794–1803.
Thiele GM, Duryee MJ, Hunter CD, et al. Immunogenic and inflammatory responses to citrullinated proteins are enhanced following modification with malondialdehyde‐acetaldehyde adducts. Int Immunopharmacol 2020;83:106433.
Thiele GM, Duryee MJ, Anderson DR, et al. Malondialdehyde‐acetaldehyde adducts and anti‐malondialdehyde‐acetaldehyde antibodies in rheumatoid arthritis. Arthritis Rheumatol 2015;67(3):645–655.
Poole JA, Mikuls TR, Thiele GM, et al. Increased susceptibility to organic dust exposure‐induced inflammatory lung disease with enhanced rheumatoid arthritis‐associated autoantigen expression in HLA‐DR4 transgenic mice. Respir Res 2022;23(1):160.
Poole JA, Thiele GM, Janike K, et al. Combined collagen‐induced arthritis and organic dust‐induced airway inflammation to model inflammatory lung disease in rheumatoid arthritis. J Bone Miner Res 2019;34(9):1733–1743.
Mikuls TR, Gaurav R, Thiele GM, et al. The impact of airborne endotoxin exposure on rheumatoid arthritis‐related joint damage, autoantigen expression, autoimmunity, and lung disease. Int Immunopharmacol 2021;100:108069.
Mikuls TR, Reimold A, Kerr GS, Cannon GW. Insights and implications of the VA Rheumatoid Arthritis Registry. Fed Pract 2015;32:24–29.
Arnett FC, Edworthy SM, Bloch DA, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum 1987;31(3):315–324.
Aripova N, Duryee MJ, Hunter CD, et al. Peptidyl arginine deiminase expression and macrophage polarization following stimulation with citrullinated and malondialdehyde‐acetaldehyde modified fibrinogen. Int Immunopharmacol 2022;110:109010.
Prevoo MLL, van't Hof MA, Kuper HH, et al. Modified disease activity scores that include twenty‐eight‐joint counts. Development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum 1995;38(1):44–48.
Baker JF, England BR, Mikuls TR, et al. Obesity, weight loss, and progression of disability in rheumatoid arthritis. Arthritis Care Res (Hoboken) 2018;70(12):1740–1747.
Cannon GW, Mikuls TR, Hayden CL, et al. Merging Veterans Affairs rheumatoid arthritis registry and pharmacy data to assess methotrexate adherence and disease activity in clinical practice. Arthritis Care Res (Hoboken) 2011;63(12):1680–1690.
England BR, Roul P, Mahajan TD, et al. Performance of administrative algorithms to identify interstitial lung disease in rheumatoid arthritis. Arthritis Care Res (Hoboken) 2020;72(10):1392–1403.
Brooks R, Baker JF, Yang Y, et al. The impact of disease severity measures on survival in U.S. veterans with rheumatoid arthritis‐associated interstitial lung disease. Rheumatology (Oxford) 2022;61(12):4667–4677.
Giles JT, Danoff SK, Sokolove J, et al. Association of fine specificity and repertoire expansion of anticitrullinated peptide antibodies with rheumatoid arthritis associated interstitial lung disease. Ann Rheum Dis 2014;73(8):1487–1494.
Kronzer VL, Hayashi K, Yoshida K, et al. Autoantibodies against citrullinated and native proteins and prediction of rheumatoid arthritis‐associated interstitial lung disease: A nested case‐control study. Lancet Rheumatol 2023;5(2):e77–e87.
Aripova N, Duryee MJ, England BR, et al. Citrullinated and malondialdehyde‐acetaldehyde modified fibrinogen activates macrophages and promotes an aggressive synovial fibroblast phenotype in patients with rheumatoid arthritis. Front Immunol 2023;14:1203548.
Thiele GM, Duryee MJ, Willis MS, et al. Malondialdehyde‐acetaldehyde (MAA) modified proteins induce pro‐inflammatory and pro‐fibrotic responses by liver endothelial cells. Comp Hepatol 2004;3(Suppl 1):S25.
Sokolove J, Bromberg R, Deane KD, et al. Autoantibody epitope spreading in the pre‐clinical phase predicts progression to rheumatoid arthritis. PLoS One 2012;7(5):e35296.
Mikuls TR, Edison J, Meeshaw E, et al. Autoantibodies to malondialdehyde‐acetaldehyde are detected prior to rheumatoid arthritis diagnosis and after other disease specific autoantibodies. Arthritis Rheumatol 2020;72(12):2025–2029.
Duryee MJ, Klassen LW, Schaffert CS, et al. Malondialdehyde‐acetaldehyde adduct is the dominant epitope after MDA modification of proteins in atherosclerosis. Free Radic Biol Med 2010;49(10):1480–1486.
Duryee MJ, Ahmad R, Eichele DD, et al. Identification of immunoglobulin G autoantibody against malondialdehyde‐acetaldehyde adducts as a novel serological biomarker for ulcerative colitis. Clin Transl Gastroenterol 2022;13(4):e00469.
معلومات مُعتمدة: IK2 CX002203 United States CX CSRD VA; PR200793 U.S. Department of Defense; P20GM130423 United States GM NIGMS NIH HHS; BX004600 Veterans Affairs Biomedical Laboratory Research and Development; IK CX002351 Veterans Affairs Clinical Science Research and Development; Rheumatology Research Foundation; U54GM115458 United States GM NIGMS NIH HHS; P20 GM130423 United States GM NIGMS NIH HHS
تواريخ الأحداث: Date Created: 20240520 Latest Revision: 20240620
رمز التحديث: 20240620
DOI: 10.1002/art.42916
PMID: 38766737
قاعدة البيانات: MEDLINE
الوصف
تدمد:2326-5205
DOI:10.1002/art.42916