Skip to main content
Top
Published in: Clinical Reviews in Allergy & Immunology 3/2020

01-12-2020 | Systemic Lupus Erythematosus

Autoimmunity in 2019

Author: Carlo Selmi

Published in: Clinical Reviews in Allergy & Immunology | Issue 3/2020

Login to get access

Abstract

Based on the PubMed data, we have been performing a yearly evaluation of the publications related to autoimmune diseases and immunology to ascertain the relative weight of the former in the scientific literature. It is particularly intriguing to observe that despite the numerous new avenues of immune-related mechanisms, such as cancer immunotherapy, the proportion of immunology manuscripts related to autoimmunity continues to increase and has been approaching 20% in 2019. As in the previous 13 years, we performed an arbitrary selection of the peer-reviewed articles published by the major dedicated Journals and discussed the common themes which continue to outnumber peculiarites in autoimmune diseases. The investigated areas included systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriatic arthritis (PsA), autoantibodies (autoAbs), and common therapeutic avenues and novel pathogenic mechanisms for autoimmune conditions. Some examples include new pathogenetic evidence which is well represented by IL21 or P2X7 receptor (P2X7R) in SLE or the application of single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq), and flow cytometry for the analysis of different cellular populations in RA. Cumulatively and of interest to the clinicians, a large number of findings continue to underline the importance of a strict relationship between basic and clinical science to define new pathogenetic and therapeutic developments. The therapeutic pipeline in autoimmunity continues to grow and maintain a constant flow of new molecules, as well illustrated in RA and PsA, and this is most certainly derived from the new basic evidence and the high-throughput tools applied to autoimmune diseases.
Literature
1.
go back to reference Zanatta E, Famoso G, Boscain F, Montisci R, Pigatto E, Polito P et al (2019) Nailfold avascular score and coronary microvascular dysfunction in systemic sclerosis: a newsworthy association. Autoimmun Rev 18:177–183PubMedCrossRef Zanatta E, Famoso G, Boscain F, Montisci R, Pigatto E, Polito P et al (2019) Nailfold avascular score and coronary microvascular dysfunction in systemic sclerosis: a newsworthy association. Autoimmun Rev 18:177–183PubMedCrossRef
2.
go back to reference Yao Q, Song Z, Wang B, Zhang JA, Mu K (2019) Thyroid disorders in patients with systemic sclerosis: a systematic review and meta-analysis. Autoimmun Rev 18:634–636PubMedCrossRef Yao Q, Song Z, Wang B, Zhang JA, Mu K (2019) Thyroid disorders in patients with systemic sclerosis: a systematic review and meta-analysis. Autoimmun Rev 18:634–636PubMedCrossRef
3.
4.
go back to reference Smith V, Vanhaecke A, Herrick AL, Distler O, Guerra MG, Denton CP et al (2019) Fast track algorithm: how to differentiate a "scleroderma pattern" from a "non-scleroderma pattern". Autoimmun Rev 18:102394PubMedCrossRef Smith V, Vanhaecke A, Herrick AL, Distler O, Guerra MG, Denton CP et al (2019) Fast track algorithm: how to differentiate a "scleroderma pattern" from a "non-scleroderma pattern". Autoimmun Rev 18:102394PubMedCrossRef
5.
go back to reference Sciascia S, Cecchi I, Massara C, Rossi D, Radin M, Ladehesa PL et al (2020) Thermography in systemic sclerosis patients and other rheumatic diseases: diagnosis, disease activity assessment, and therapeutic monitoring. Autoimmun Rev 19:102449PubMedCrossRef Sciascia S, Cecchi I, Massara C, Rossi D, Radin M, Ladehesa PL et al (2020) Thermography in systemic sclerosis patients and other rheumatic diseases: diagnosis, disease activity assessment, and therapeutic monitoring. Autoimmun Rev 19:102449PubMedCrossRef
6.
go back to reference Riccardi A, Marcoccia A, Borgia A, Guastafierro T, Bondanini F, Fasano S et al (2019) Undifferentiated connective tissue disease at risk of systemic sclerosis: a weighted score to identify patients who will evolve. Autoimmun Rev 18:102358PubMedCrossRef Riccardi A, Marcoccia A, Borgia A, Guastafierro T, Bondanini F, Fasano S et al (2019) Undifferentiated connective tissue disease at risk of systemic sclerosis: a weighted score to identify patients who will evolve. Autoimmun Rev 18:102358PubMedCrossRef
7.
go back to reference Narvaez J, Pirola JP, LLuch J, Juarez P, Nolla JM, Valenzuela A (2019) Effectiveness and safety of rituximab for the treatment of refractory systemic sclerosis associated calcinosis: a case series and systematic review of the literature. Autoimmun Rev 18:262–269PubMedCrossRef Narvaez J, Pirola JP, LLuch J, Juarez P, Nolla JM, Valenzuela A (2019) Effectiveness and safety of rituximab for the treatment of refractory systemic sclerosis associated calcinosis: a case series and systematic review of the literature. Autoimmun Rev 18:262–269PubMedCrossRef
8.
go back to reference Napodano C, Pocino K, Rigante D, Stefanile A, Gulli F, Marino M et al (2019) Free light chains and autoimmunity. Autoimmun Rev 18:484–492PubMedCrossRef Napodano C, Pocino K, Rigante D, Stefanile A, Gulli F, Marino M et al (2019) Free light chains and autoimmunity. Autoimmun Rev 18:484–492PubMedCrossRef
9.
go back to reference Molano-Gonzalez N, Rojas M, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Rodriguez Y et al (2019) Cluster analysis of autoimmune rheumatic diseases based on autoantibodies. New insights for polyautoimmunity. J Autoimmun 98:24–32PubMedCrossRef Molano-Gonzalez N, Rojas M, Monsalve DM, Pacheco Y, Acosta-Ampudia Y, Rodriguez Y et al (2019) Cluster analysis of autoimmune rheumatic diseases based on autoantibodies. New insights for polyautoimmunity. J Autoimmun 98:24–32PubMedCrossRef
10.
go back to reference Meridor K, Levy Y (2020) Systemic sclerosis induced by CNS stimulants for ADHD: a case series and review of the literature. Autoimmun Rev 19:102439PubMedCrossRef Meridor K, Levy Y (2020) Systemic sclerosis induced by CNS stimulants for ADHD: a case series and review of the literature. Autoimmun Rev 19:102439PubMedCrossRef
11.
12.
go back to reference Jafarinejad-Farsangi S, Gharibdoost F, Farazmand A, Kavosi H, Jamshidi A, Karimizadeh E et al (2019) MicroRNA-21 and microRNA-29a modulate the expression of collagen in dermal fibroblasts of patients with systemic sclerosis. Autoimmunity 52:108–116PubMedCrossRef Jafarinejad-Farsangi S, Gharibdoost F, Farazmand A, Kavosi H, Jamshidi A, Karimizadeh E et al (2019) MicroRNA-21 and microRNA-29a modulate the expression of collagen in dermal fibroblasts of patients with systemic sclerosis. Autoimmunity 52:108–116PubMedCrossRef
13.
go back to reference Henry TW, Mendoza FA, Jimenez SA (2019) Role of microRNA in the pathogenesis of systemic sclerosis tissue fibrosis and vasculopathy. Autoimmun Rev 18:102396PubMedCrossRef Henry TW, Mendoza FA, Jimenez SA (2019) Role of microRNA in the pathogenesis of systemic sclerosis tissue fibrosis and vasculopathy. Autoimmun Rev 18:102396PubMedCrossRef
14.
go back to reference Frantz C, Huscher D, Avouac J, Hachulla E, Balbir-Gurman A, Riemekasten G et al (2020) Outcomes of limited cutaneous systemic sclerosis patients: results on more than 12,000 patients from the EUSTAR database. Autoimmun Rev 19:102452PubMedCrossRef Frantz C, Huscher D, Avouac J, Hachulla E, Balbir-Gurman A, Riemekasten G et al (2020) Outcomes of limited cutaneous systemic sclerosis patients: results on more than 12,000 patients from the EUSTAR database. Autoimmun Rev 19:102452PubMedCrossRef
15.
go back to reference Chaigne B, Rodeia S, Benmostefa N, Berezne A, Authier J, Cohen P et al (2020) Corticosteroid-sparing benefit of intravenous immunoglobulin in systemic sclerosis-associated myopathy: a comparative study in 52 patients. Autoimmun Rev 19:102431PubMedCrossRef Chaigne B, Rodeia S, Benmostefa N, Berezne A, Authier J, Cohen P et al (2020) Corticosteroid-sparing benefit of intravenous immunoglobulin in systemic sclerosis-associated myopathy: a comparative study in 52 patients. Autoimmun Rev 19:102431PubMedCrossRef
16.
go back to reference Cattalini M, Soliani M, Caparello MC, Cimaz R (2019) Sex differences in pediatric rheumatology. Clin Rev Allergy Immunol 56:293–307PubMedCrossRef Cattalini M, Soliani M, Caparello MC, Cimaz R (2019) Sex differences in pediatric rheumatology. Clin Rev Allergy Immunol 56:293–307PubMedCrossRef
17.
go back to reference Blagojevic J, Legendre P, Matucci-Cerinic M, Mouthon L (2019) Is there today a place for corticosteroids in the treatment of scleroderma? Autoimmun Rev 18:102403PubMedCrossRef Blagojevic J, Legendre P, Matucci-Cerinic M, Mouthon L (2019) Is there today a place for corticosteroids in the treatment of scleroderma? Autoimmun Rev 18:102403PubMedCrossRef
18.
go back to reference Bellan M, Patrucco F, Barone-Adesi F, Gavelli F, Castello LM, Nerviani A et al (2020) Targeting CD20 in the treatment of interstitial lung diseases related to connective tissue diseases: a systematic review. Autoimmun Rev 19:102453PubMedCrossRef Bellan M, Patrucco F, Barone-Adesi F, Gavelli F, Castello LM, Nerviani A et al (2020) Targeting CD20 in the treatment of interstitial lung diseases related to connective tissue diseases: a systematic review. Autoimmun Rev 19:102453PubMedCrossRef
19.
20.
go back to reference Lescoat A, Ballerie A, Jouneau S, Fardel O, Vernhet L, Jego P et al (2019) M1/M2 polarisation state of M-CSF blood-derived macrophages in systemic sclerosis. Ann Rheum Dis 78:e127PubMedCrossRef Lescoat A, Ballerie A, Jouneau S, Fardel O, Vernhet L, Jego P et al (2019) M1/M2 polarisation state of M-CSF blood-derived macrophages in systemic sclerosis. Ann Rheum Dis 78:e127PubMedCrossRef
21.
go back to reference Skaug B, Khanna D, Swindell WR, Hinchcliff ME, Frech TM, Steen VD et al (2020) Global skin gene expression analysis of early diffuse cutaneous systemic sclerosis shows a prominent innate and adaptive inflammatory profile. Ann Rheum Dis 79:379–386PubMedCrossRef Skaug B, Khanna D, Swindell WR, Hinchcliff ME, Frech TM, Steen VD et al (2020) Global skin gene expression analysis of early diffuse cutaneous systemic sclerosis shows a prominent innate and adaptive inflammatory profile. Ann Rheum Dis 79:379–386PubMedCrossRef
22.
go back to reference Skarstein K, Jensen JL, Galtung H, Jonsson R, Brokstad K, Aqrawi LA (2019) Autoantigen-specific B cells and plasma cells are prominent in areas of fatty infiltration in salivary glands of patients with primary Sjogren's syndrome. Autoimmunity 52:242–250PubMedCrossRef Skarstein K, Jensen JL, Galtung H, Jonsson R, Brokstad K, Aqrawi LA (2019) Autoantigen-specific B cells and plasma cells are prominent in areas of fatty infiltration in salivary glands of patients with primary Sjogren's syndrome. Autoimmunity 52:242–250PubMedCrossRef
23.
go back to reference Shin JI, Lee KH, Joo YH, Lee JM, Jeon J, Jung HJ et al (2019) Inflammasomes and autoimmune and rheumatic diseases: a comprehensive review. J Autoimmun 103:102299PubMedCrossRef Shin JI, Lee KH, Joo YH, Lee JM, Jeon J, Jung HJ et al (2019) Inflammasomes and autoimmune and rheumatic diseases: a comprehensive review. J Autoimmun 103:102299PubMedCrossRef
24.
go back to reference Martin-Nares E, Hernandez-Molina G (2019) Novel autoantibodies in Sjogren's syndrome: a comprehensive review. Autoimmun Rev 18:192–198PubMedCrossRef Martin-Nares E, Hernandez-Molina G (2019) Novel autoantibodies in Sjogren's syndrome: a comprehensive review. Autoimmun Rev 18:192–198PubMedCrossRef
25.
go back to reference Malagon C, Gomez MDP, Mosquera C, Vargas C, Gonzalez T, Arango C et al (2019) Juvenile polyautoimmunity in a rheumatology setting. Autoimmun Rev 18:369–381PubMedCrossRef Malagon C, Gomez MDP, Mosquera C, Vargas C, Gonzalez T, Arango C et al (2019) Juvenile polyautoimmunity in a rheumatology setting. Autoimmun Rev 18:369–381PubMedCrossRef
26.
go back to reference Long D, Chen Y, Wu H, Zhao M, Lu Q (2019) Clinical significance and immunobiology of IL-21 in autoimmunity. J Autoimmun 99:1–14PubMedCrossRef Long D, Chen Y, Wu H, Zhao M, Lu Q (2019) Clinical significance and immunobiology of IL-21 in autoimmunity. J Autoimmun 99:1–14PubMedCrossRef
27.
go back to reference Huang C, Zhu HX, Yao Y, Bian ZH, Zheng YJ, Li L et al (2019) Immune checkpoint molecules Possible future therapeutic implications in autoimmune diseases. J Autoimmun 104:102333PubMedCrossRef Huang C, Zhu HX, Yao Y, Bian ZH, Zheng YJ, Li L et al (2019) Immune checkpoint molecules Possible future therapeutic implications in autoimmune diseases. J Autoimmun 104:102333PubMedCrossRef
28.
go back to reference Ferrari SM, Fallahi P, Ruffilli I, Elia G, Ragusa F, Benvenga S et al (2019) The association of other autoimmune diseases in patients with Graves' disease (with or without ophthalmopathy): review of the literature and report of a large series. Autoimmun Rev 18:287–292PubMedCrossRef Ferrari SM, Fallahi P, Ruffilli I, Elia G, Ragusa F, Benvenga S et al (2019) The association of other autoimmune diseases in patients with Graves' disease (with or without ophthalmopathy): review of the literature and report of a large series. Autoimmun Rev 18:287–292PubMedCrossRef
29.
go back to reference Clancy RM, Marion MC, Ainsworth HC, Blaser MJ, Chang M, Howard TD et al (2020) Salivary dysbiosis and the clinical spectrum in anti-Ro positive mothers of children with neonatal lupus. J Autoimmun 107:102354PubMedCrossRef Clancy RM, Marion MC, Ainsworth HC, Blaser MJ, Chang M, Howard TD et al (2020) Salivary dysbiosis and the clinical spectrum in anti-Ro positive mothers of children with neonatal lupus. J Autoimmun 107:102354PubMedCrossRef
30.
go back to reference Clancy RM, Halushka M, Rasmussen SE, Lhakhang T, Chang M, Buyon JP (2019) Siglec-1 macrophages and the contribution of IFN to the development of autoimmune congenital heart block. J Immunol 202:48–55PubMedCrossRef Clancy RM, Halushka M, Rasmussen SE, Lhakhang T, Chang M, Buyon JP (2019) Siglec-1 macrophages and the contribution of IFN to the development of autoimmune congenital heart block. J Immunol 202:48–55PubMedCrossRef
31.
go back to reference Applbaum E, Lichtbroun A (2019) Novel Sjogren's autoantibodies found in fibromyalgia patients with sicca and/or xerostomia. Autoimmun Rev 18:199–202PubMedCrossRef Applbaum E, Lichtbroun A (2019) Novel Sjogren's autoantibodies found in fibromyalgia patients with sicca and/or xerostomia. Autoimmun Rev 18:199–202PubMedCrossRef
32.
go back to reference Anaya JM, Restrepo-Jimenez P, Rodriguez Y, Rodriguez-Jimenez M, Acosta-Ampudia Y, Monsalve DM et al (2019) Sjogren's syndrome and autoimmune thyroid disease: two sides of the same coin. Clin Rev Allergy Immunol 56:362–374PubMedCrossRef Anaya JM, Restrepo-Jimenez P, Rodriguez Y, Rodriguez-Jimenez M, Acosta-Ampudia Y, Monsalve DM et al (2019) Sjogren's syndrome and autoimmune thyroid disease: two sides of the same coin. Clin Rev Allergy Immunol 56:362–374PubMedCrossRef
33.
go back to reference Dorner T, Posch MG, Li Y, Petricoul O, Cabanski M, Milojevic JM et al (2019) Treatment of primary Sjogren's syndrome with ianalumab (VAY736) targeting B cells by BAFF receptor blockade coupled with enhanced, antibody-dependent cellular cytotoxicity. Ann Rheum Dis 78:641–647PubMedCrossRef Dorner T, Posch MG, Li Y, Petricoul O, Cabanski M, Milojevic JM et al (2019) Treatment of primary Sjogren's syndrome with ianalumab (VAY736) targeting B cells by BAFF receptor blockade coupled with enhanced, antibody-dependent cellular cytotoxicity. Ann Rheum Dis 78:641–647PubMedCrossRef
34.
go back to reference Ramos-Casals M, Brito-Zeron P, Bombardieri S, Bootsma H, De Vita S, Dorner T et al (2020) EULAR recommendations for the management of Sjogren's syndrome with topical and systemic therapies. Ann Rheum Dis 79:3–18PubMedCrossRef Ramos-Casals M, Brito-Zeron P, Bombardieri S, Bootsma H, De Vita S, Dorner T et al (2020) EULAR recommendations for the management of Sjogren's syndrome with topical and systemic therapies. Ann Rheum Dis 79:3–18PubMedCrossRef
35.
go back to reference van der Meulen TA, van Nimwegen JF, Harmsen HJM, Liefers SC, van der Tuuk K, Kroese FGM et al (2019) Normal vaginal microbiome in women with primary Sjogren's syndrome-associated vaginal dryness. Ann Rheum Dis 78:707–709PubMedCrossRef van der Meulen TA, van Nimwegen JF, Harmsen HJM, Liefers SC, van der Tuuk K, Kroese FGM et al (2019) Normal vaginal microbiome in women with primary Sjogren's syndrome-associated vaginal dryness. Ann Rheum Dis 78:707–709PubMedCrossRef
36.
go back to reference Lee J, Baek S, Koh JH, Kim JW, Kim SY, Chung SH et al (2019) Soluble siglec-5 is a novel salivary biomarker for primary Sjogren's syndrome. J Autoimmun 100:114–119PubMedCrossRef Lee J, Baek S, Koh JH, Kim JW, Kim SY, Chung SH et al (2019) Soluble siglec-5 is a novel salivary biomarker for primary Sjogren's syndrome. J Autoimmun 100:114–119PubMedCrossRef
37.
go back to reference Wu X, Liu Y, Wei W, Liu ML (2019) Extracellular vesicles in autoimmune vasculitis - little dirts light the fire in blood vessels. Autoimmun Rev 18:593–606PubMedPubMedCentralCrossRef Wu X, Liu Y, Wei W, Liu ML (2019) Extracellular vesicles in autoimmune vasculitis - little dirts light the fire in blood vessels. Autoimmun Rev 18:593–606PubMedPubMedCentralCrossRef
38.
go back to reference Watanabe-Kusunoki K, Nakazawa D, Kusunoki Y, Kudo T, Hattanda F, Nishio S et al (2020) Recombinant thrombomodulin ameliorates autoimmune vasculitis via immune response regulation and tissue injury protection. J Autoimmun 108:102390PubMedCrossRef Watanabe-Kusunoki K, Nakazawa D, Kusunoki Y, Kudo T, Hattanda F, Nishio S et al (2020) Recombinant thrombomodulin ameliorates autoimmune vasculitis via immune response regulation and tissue injury protection. J Autoimmun 108:102390PubMedCrossRef
39.
go back to reference Wang C, Deng H, Gong Y, You R, Chen M, Zhao MH (2020) Effect of high mobility group box 1 on toll-like receptor 9 in B cells in myeloperoxidase-ANCA-associated vasculitis. Autoimmunity 53:28–34PubMedCrossRef Wang C, Deng H, Gong Y, You R, Chen M, Zhao MH (2020) Effect of high mobility group box 1 on toll-like receptor 9 in B cells in myeloperoxidase-ANCA-associated vasculitis. Autoimmunity 53:28–34PubMedCrossRef
40.
go back to reference Walls CA, Basu N, Hutcheon G, Erwig LP, Little MA, Kidder D (2020) A novel 4-dimensional live-cell imaging system to study leukocyte-endothelial dynamics in ANCA-associated vasculitis. Autoimmunity 53:148–155PubMedCrossRef Walls CA, Basu N, Hutcheon G, Erwig LP, Little MA, Kidder D (2020) A novel 4-dimensional live-cell imaging system to study leukocyte-endothelial dynamics in ANCA-associated vasculitis. Autoimmunity 53:148–155PubMedCrossRef
41.
go back to reference Tahvildari M, Dana R (2019) Low-dose IL-2 therapy in transplantation, autoimmunity, and inflammatory diseases. J Immunol 203:2749–2755PubMedCrossRef Tahvildari M, Dana R (2019) Low-dose IL-2 therapy in transplantation, autoimmunity, and inflammatory diseases. J Immunol 203:2749–2755PubMedCrossRef
42.
go back to reference Hamano Y, Ito F, Suzuki O, Koura M, Matsuoka S, Kobayashi T et al (2019) Vasculitis and crescentic glomerulonephritis in a newly established congenic mouse strain derived from ANCA-associated vasculitis-prone SCG/Kj mice. Autoimmunity 52:208–219PubMedCrossRef Hamano Y, Ito F, Suzuki O, Koura M, Matsuoka S, Kobayashi T et al (2019) Vasculitis and crescentic glomerulonephritis in a newly established congenic mouse strain derived from ANCA-associated vasculitis-prone SCG/Kj mice. Autoimmunity 52:208–219PubMedCrossRef
43.
go back to reference Frangou E, Vassilopoulos D, Boletis J, Boumpas DT (2019) An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): implications for the pathogenesis and treatment. Autoimmun Rev 18:751–760PubMedCrossRef Frangou E, Vassilopoulos D, Boletis J, Boumpas DT (2019) An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): implications for the pathogenesis and treatment. Autoimmun Rev 18:751–760PubMedCrossRef
44.
go back to reference Enocsson H, Wirestam L, Dahle C, Padyukov L, Jonsen A, Urowitz MB et al (2020) Soluble urokinase plasminogen activator receptor (suPAR) levels predict damage accrual in patients with recent-onset systemic lupus erythematosus. J Autoimmun 106:102340PubMedCrossRef Enocsson H, Wirestam L, Dahle C, Padyukov L, Jonsen A, Urowitz MB et al (2020) Soluble urokinase plasminogen activator receptor (suPAR) levels predict damage accrual in patients with recent-onset systemic lupus erythematosus. J Autoimmun 106:102340PubMedCrossRef
45.
go back to reference Dolff S, Witzke O, Wilde B (2019) Th17 cells in renal inflammation and autoimmunity. Autoimmun Rev 18:129–136PubMedCrossRef Dolff S, Witzke O, Wilde B (2019) Th17 cells in renal inflammation and autoimmunity. Autoimmun Rev 18:129–136PubMedCrossRef
46.
go back to reference Klapa S, Muller A, Koch A, Heidecke H, Kahler W, Junker J et al (2019) Decreased endothelin receptor A autoantibody levels are associated with early ischaemic events in patients with giant-cell arteritis. Ann Rheum Dis 78:1443–1444PubMedCrossRef Klapa S, Muller A, Koch A, Heidecke H, Kahler W, Junker J et al (2019) Decreased endothelin receptor A autoantibody levels are associated with early ischaemic events in patients with giant-cell arteritis. Ann Rheum Dis 78:1443–1444PubMedCrossRef
47.
go back to reference Kong X, Sawalha AH (2019) Takayasu arteritis risk locus in IL6 represses the anti-inflammatory gene GPNMB through chromatin looping and recruiting MEF2-HDAC complex. Ann Rheum Dis 78:1388–1397PubMedCrossRef Kong X, Sawalha AH (2019) Takayasu arteritis risk locus in IL6 represses the anti-inflammatory gene GPNMB through chromatin looping and recruiting MEF2-HDAC complex. Ann Rheum Dis 78:1388–1397PubMedCrossRef
48.
go back to reference Michailidou D, Rosenblum JS, Rimland CA, Marko J, Ahlman MA, Grayson PC (2020) Clinical symptoms and associated vascular imaging findings in Takayasu's arteritis compared to giant cell arteritis. Ann Rheum Dis 79:262–267PubMedCrossRef Michailidou D, Rosenblum JS, Rimland CA, Marko J, Ahlman MA, Grayson PC (2020) Clinical symptoms and associated vascular imaging findings in Takayasu's arteritis compared to giant cell arteritis. Ann Rheum Dis 79:262–267PubMedCrossRef
49.
go back to reference Vandergheynst FA, Lelubre C (2019) Mycophenolate mofetil: a step forward in the induction treatment of ANCA-associated vasculitis? Comment on the article by Jones et al. Annals of the rheumatic diseases Vandergheynst FA, Lelubre C (2019) Mycophenolate mofetil: a step forward in the induction treatment of ANCA-associated vasculitis? Comment on the article by Jones et al. Annals of the rheumatic diseases
50.
go back to reference Dieudonne Y, Gies V, Guffroy A, Keime C, Bird AK, Liesveld J et al (2019) Transitional B cells in quiescent SLE: an early checkpoint imprinted by IFN. J Autoimmun 102:150–158PubMedPubMedCentralCrossRef Dieudonne Y, Gies V, Guffroy A, Keime C, Bird AK, Liesveld J et al (2019) Transitional B cells in quiescent SLE: an early checkpoint imprinted by IFN. J Autoimmun 102:150–158PubMedPubMedCentralCrossRef
51.
go back to reference Pisetsky DS (2020) Evolving story of autoantibodies in systemic lupus erythematosus. J Autoimmun 110:102356PubMedCrossRef Pisetsky DS (2020) Evolving story of autoantibodies in systemic lupus erythematosus. J Autoimmun 110:102356PubMedCrossRef
52.
go back to reference Franks SE, Getahun A, Cambier JC (2019) A precision B cell-targeted therapeutic approach to autoimmunity caused by phosphatidylinositol 3-kinase pathway dysregulation. J Immunol 202:3381–3393PubMedCrossRef Franks SE, Getahun A, Cambier JC (2019) A precision B cell-targeted therapeutic approach to autoimmunity caused by phosphatidylinositol 3-kinase pathway dysregulation. J Immunol 202:3381–3393PubMedCrossRef
53.
go back to reference Feng Y, Yang M, Wu H, Lu Q (2020) The pathological role of B cells in systemic lupus erythematosus: from basic research to clinical. Autoimmunity 53:56–64PubMedCrossRef Feng Y, Yang M, Wu H, Lu Q (2020) The pathological role of B cells in systemic lupus erythematosus: from basic research to clinical. Autoimmunity 53:56–64PubMedCrossRef
54.
go back to reference Cao F, Hu LQ, Yao SR, Hu Y, Wang DG, Fan YG et al (2019) P2X7 receptor: a potential therapeutic target for autoimmune diseases. Autoimmun Rev 18:767–777PubMedCrossRef Cao F, Hu LQ, Yao SR, Hu Y, Wang DG, Fan YG et al (2019) P2X7 receptor: a potential therapeutic target for autoimmune diseases. Autoimmun Rev 18:767–777PubMedCrossRef
55.
go back to reference Pernis AB, Ivashkiv LB (2019) ‘-Omics’ shed light on B cells in lupus. Nat Immunol 20:946–948PubMedCrossRef Pernis AB, Ivashkiv LB (2019) ‘-Omics’ shed light on B cells in lupus. Nat Immunol 20:946–948PubMedCrossRef
56.
go back to reference Cunningham MA, Richard ML, Wirth JR, Scott JL, Eudaly J, Ruiz P et al (2019) Novel mechanism for estrogen receptor alpha modulation of murine lupus. J Autoimmun 97:59–69PubMedCrossRef Cunningham MA, Richard ML, Wirth JR, Scott JL, Eudaly J, Ruiz P et al (2019) Novel mechanism for estrogen receptor alpha modulation of murine lupus. J Autoimmun 97:59–69PubMedCrossRef
57.
go back to reference Syrett CM, Sierra I, Beethem ZT, Dubin AH, Anguera MC (2020) Loss of epigenetic modifications on the inactive X chromosome and sex-biased gene expression profiles in B cells from NZB/W F1 mice with lupus-like disease. J Autoimmun 107:102357PubMedCrossRef Syrett CM, Sierra I, Beethem ZT, Dubin AH, Anguera MC (2020) Loss of epigenetic modifications on the inactive X chromosome and sex-biased gene expression profiles in B cells from NZB/W F1 mice with lupus-like disease. J Autoimmun 107:102357PubMedCrossRef
58.
go back to reference Lownik JC, Wimberly JL, Conrad DH, Martin RK (2019) B cell ADAM10 controls murine lupus progression through regulation of the ICOS:ICOS ligand axis. J Immunol 202:664–674PubMedCrossRef Lownik JC, Wimberly JL, Conrad DH, Martin RK (2019) B cell ADAM10 controls murine lupus progression through regulation of the ICOS:ICOS ligand axis. J Immunol 202:664–674PubMedCrossRef
59.
go back to reference Giltiay NV, Giordano D, Clark EA (2019) The plasticity of newly formed B cells. J Immunol 203:3095–3104PubMedCrossRef Giltiay NV, Giordano D, Clark EA (2019) The plasticity of newly formed B cells. J Immunol 203:3095–3104PubMedCrossRef
60.
go back to reference Liu X, Zhang W, Zhao M, Fu L, Liu L, Wu J et al (2019) T cell receptor beta repertoires as novel diagnostic markers for systemic lupus erythematosus and rheumatoid arthritis. Ann Rheum Dis 78:1070–1078PubMedCrossRef Liu X, Zhang W, Zhao M, Fu L, Liu L, Wu J et al (2019) T cell receptor beta repertoires as novel diagnostic markers for systemic lupus erythematosus and rheumatoid arthritis. Ann Rheum Dis 78:1070–1078PubMedCrossRef
61.
go back to reference Frangou E, Chrysanthopoulou A, Mitsios A, Kambas K, Arelaki S, Angelidou I et al (2019) REDD1/autophagy pathway promotes thromboinflammation and fibrosis in human systemic lupus erythematosus (SLE) through NETs decorated with tissue factor (TF) and interleukin-17A (IL-17A). Ann Rheum Dis 78:238–248PubMedCrossRef Frangou E, Chrysanthopoulou A, Mitsios A, Kambas K, Arelaki S, Angelidou I et al (2019) REDD1/autophagy pathway promotes thromboinflammation and fibrosis in human systemic lupus erythematosus (SLE) through NETs decorated with tissue factor (TF) and interleukin-17A (IL-17A). Ann Rheum Dis 78:238–248PubMedCrossRef
62.
go back to reference Linge P, Arve S, Olsson LM, Leonard D, Sjowall C, Frodlund M et al (2020) NCF1-339 polymorphism is associated with altered formation of neutrophil extracellular traps, high serum interferon activity and antiphospholipid syndrome in systemic lupus erythematosus. Ann Rheum Dis 79:254–261PubMedCrossRef Linge P, Arve S, Olsson LM, Leonard D, Sjowall C, Frodlund M et al (2020) NCF1-339 polymorphism is associated with altered formation of neutrophil extracellular traps, high serum interferon activity and antiphospholipid syndrome in systemic lupus erythematosus. Ann Rheum Dis 79:254–261PubMedCrossRef
63.
go back to reference Odqvist L, Jevnikar Z, Riise R, Oberg L, Rhedin M, Leonard D et al (2019) Genetic variations in A20 DUB domain provide a genetic link to citrullination and neutrophil extracellular traps in systemic lupus erythematosus. Ann Rheum Dis 78:1363–1370PubMedCrossRef Odqvist L, Jevnikar Z, Riise R, Oberg L, Rhedin M, Leonard D et al (2019) Genetic variations in A20 DUB domain provide a genetic link to citrullination and neutrophil extracellular traps in systemic lupus erythematosus. Ann Rheum Dis 78:1363–1370PubMedCrossRef
64.
go back to reference Bach M, Moon J, Moore R, Pan T, Nelson JL, Lood C (2020) A neutrophil activation biomarker panel in prognosis and monitoring of patients with rheumatoid arthritis. Arthritis Rheum 72:47–56CrossRef Bach M, Moon J, Moore R, Pan T, Nelson JL, Lood C (2020) A neutrophil activation biomarker panel in prognosis and monitoring of patients with rheumatoid arthritis. Arthritis Rheum 72:47–56CrossRef
65.
go back to reference Curran CS, Gupta S, Sanz I, Sharon E (2019) PD-1 immunobiology in systemic lupus erythematosus. J Autoimmun 97:1–9PubMedCrossRef Curran CS, Gupta S, Sanz I, Sharon E (2019) PD-1 immunobiology in systemic lupus erythematosus. J Autoimmun 97:1–9PubMedCrossRef
66.
go back to reference Horuluoglu B, Bayik D, Kayraklioglu N, Goguet E, Kaplan MJ, Klinman DM (2019) PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J Autoimmun 99:24–32PubMedPubMedCentralCrossRef Horuluoglu B, Bayik D, Kayraklioglu N, Goguet E, Kaplan MJ, Klinman DM (2019) PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J Autoimmun 99:24–32PubMedPubMedCentralCrossRef
67.
go back to reference Panousis NI, Bertsias GK, Ongen H, Gergianaki I, Tektonidou MG, Trachana M et al (2019) Combined genetic and transcriptome analysis of patients with SLE: distinct, targetable signatures for susceptibility and severity. Ann Rheum Dis 78:1079–1089PubMedCrossRef Panousis NI, Bertsias GK, Ongen H, Gergianaki I, Tektonidou MG, Trachana M et al (2019) Combined genetic and transcriptome analysis of patients with SLE: distinct, targetable signatures for susceptibility and severity. Ann Rheum Dis 78:1079–1089PubMedCrossRef
68.
go back to reference Wang B, Chen S, Zheng Q, Gao Z, Chen R, Xuan J et al. (2019) Development and initial validation of diagnostic gene signatures for systemic lupus erythematosus. Ann Rheum Dis Wang B, Chen S, Zheng Q, Gao Z, Chen R, Xuan J et al. (2019) Development and initial validation of diagnostic gene signatures for systemic lupus erythematosus. Ann Rheum Dis
69.
go back to reference Wang Y, Chen S, Chen J, Xie X, Gao S, Zhang C et al (2020) Germline genetic patterns underlying familial rheumatoid arthritis, systemic lupus erythematosus and primary Sjogren's syndrome highlight T cell-initiated autoimmunity. Ann Rheum Dis 79:268–275PubMedCrossRef Wang Y, Chen S, Chen J, Xie X, Gao S, Zhang C et al (2020) Germline genetic patterns underlying familial rheumatoid arthritis, systemic lupus erythematosus and primary Sjogren's syndrome highlight T cell-initiated autoimmunity. Ann Rheum Dis 79:268–275PubMedCrossRef
70.
go back to reference Zhang SX, Wang J, Chen JW, Zhang MX, Zhang YF, Hu FY et al. (2019) The level of peripheral regulatory T cells is linked to changes in gut commensal microflora in patients with systemic lupus erythematosus Ann Rheum Dis Zhang SX, Wang J, Chen JW, Zhang MX, Zhang YF, Hu FY et al. (2019) The level of peripheral regulatory T cells is linked to changes in gut commensal microflora in patients with systemic lupus erythematosus Ann Rheum Dis
71.
go back to reference Zhao Z, Ren J, Dai C, Kannapell CC, Wang H, Gaskin F et al (2019) Nature of T cell epitopes in lupus antigens and HLA-DR determines autoantibody initiation and diversification. Ann Rheum Dis 78:380–390PubMedCrossRef Zhao Z, Ren J, Dai C, Kannapell CC, Wang H, Gaskin F et al (2019) Nature of T cell epitopes in lupus antigens and HLA-DR determines autoantibody initiation and diversification. Ann Rheum Dis 78:380–390PubMedCrossRef
72.
go back to reference Anaya-Macias BU, De la Cruz-Mosso U, Palafox-Sanchez CA, Parra-Rojas I, Martinez-Bonilla G, Gonzalez-Lopez L et al (2020) The -675 4G/5G PAI-1 polymorphism confers genetic susceptibility to systemic lupus erythematosus, its clinical manifestations, and comorbidities in Mexican-Mestizo population. Autoimmunity 53:71–77PubMedCrossRef Anaya-Macias BU, De la Cruz-Mosso U, Palafox-Sanchez CA, Parra-Rojas I, Martinez-Bonilla G, Gonzalez-Lopez L et al (2020) The -675 4G/5G PAI-1 polymorphism confers genetic susceptibility to systemic lupus erythematosus, its clinical manifestations, and comorbidities in Mexican-Mestizo population. Autoimmunity 53:71–77PubMedCrossRef
73.
go back to reference Liu LN, Wang P, Zou YF, Xu Z, Cheng J, Zhang Y et al (2019) Semaphorin-3A, semaphorin-7A gene single nucleotide polymorphisms, and systemic lupus erythematosus susceptibility. Autoimmunity 52:161–167PubMedCrossRef Liu LN, Wang P, Zou YF, Xu Z, Cheng J, Zhang Y et al (2019) Semaphorin-3A, semaphorin-7A gene single nucleotide polymorphisms, and systemic lupus erythematosus susceptibility. Autoimmunity 52:161–167PubMedCrossRef
74.
go back to reference Murdaca G, Tonacci A, Negrini S, Greco M, Borro M, Puppo F et al (2019) Emerging role of vitamin D in autoimmune diseases: an update on evidence and therapeutic implications. Autoimmun Rev 18:102350PubMedCrossRef Murdaca G, Tonacci A, Negrini S, Greco M, Borro M, Puppo F et al (2019) Emerging role of vitamin D in autoimmune diseases: an update on evidence and therapeutic implications. Autoimmun Rev 18:102350PubMedCrossRef
75.
go back to reference Correa Freitas E, Evelyn Karnopp T, de Souza Silva JM, Cavalheiro do Espirito Santo R, da Rosa TH, de Oliveira MS et al (2019) Vitamin D supplementation ameliorates arthritis but does not alleviates renal injury in pristane-induced lupus model. Autoimmunity 52:69–77PubMedCrossRef Correa Freitas E, Evelyn Karnopp T, de Souza Silva JM, Cavalheiro do Espirito Santo R, da Rosa TH, de Oliveira MS et al (2019) Vitamin D supplementation ameliorates arthritis but does not alleviates renal injury in pristane-induced lupus model. Autoimmunity 52:69–77PubMedCrossRef
76.
go back to reference Soni C, Sinha I, Fasnacht MJ, Olsen NJ, Rahman ZSM, Sinha R (2019) Selenium supplementation suppresses immunological and serological features of lupus in B6.Sle1b mice. Autoimmunity 52:57–68PubMedCrossRefPubMedCentral Soni C, Sinha I, Fasnacht MJ, Olsen NJ, Rahman ZSM, Sinha R (2019) Selenium supplementation suppresses immunological and serological features of lupus in B6.Sle1b mice. Autoimmunity 52:57–68PubMedCrossRefPubMedCentral
77.
go back to reference Briand C, Fremond ML, Bessis D, Carbasse A, Rice GI, Bondet V et al (2019) Efficacy of JAK1/2 inhibition in the treatment of chilblain lupus due to TREX1 deficiency. Ann Rheum Dis 78:431–433PubMedCrossRef Briand C, Fremond ML, Bessis D, Carbasse A, Rice GI, Bondet V et al (2019) Efficacy of JAK1/2 inhibition in the treatment of chilblain lupus due to TREX1 deficiency. Ann Rheum Dis 78:431–433PubMedCrossRef
78.
go back to reference Goropevsek A, Holcar M, Pahor A, Avcin T (2019) STAT signaling as a marker of SLE disease severity and implications for clinical therapy. Autoimmun Rev 18:144–154PubMedCrossRef Goropevsek A, Holcar M, Pahor A, Avcin T (2019) STAT signaling as a marker of SLE disease severity and implications for clinical therapy. Autoimmun Rev 18:144–154PubMedCrossRef
79.
go back to reference Dall'Era M, Pauli ML, Remedios K, Taravati K, Sandova PM, Putnam AL et al (2019) Adoptive Treg cell therapy in a patient with systemic lupus erythematosus. Arthritis Rheum 71:431–440CrossRef Dall'Era M, Pauli ML, Remedios K, Taravati K, Sandova PM, Putnam AL et al (2019) Adoptive Treg cell therapy in a patient with systemic lupus erythematosus. Arthritis Rheum 71:431–440CrossRef
80.
go back to reference Zhang F, Wei K, Slowikowski K, Fonseka CY, Rao DA, Kelly S et al (2019) Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol 20:928–942PubMedPubMedCentralCrossRef Zhang F, Wei K, Slowikowski K, Fonseka CY, Rao DA, Kelly S et al (2019) Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol 20:928–942PubMedPubMedCentralCrossRef
81.
go back to reference Scheinecker C, Goschl L, Bonelli M (2020) Treg cells in health and autoimmune diseases: new insights from single cell analysis. J Autoimmun 110:102376PubMedCrossRef Scheinecker C, Goschl L, Bonelli M (2020) Treg cells in health and autoimmune diseases: new insights from single cell analysis. J Autoimmun 110:102376PubMedCrossRef
82.
go back to reference Cammarata I, Martire C, Citro A, Raimondo D, Fruci D, Melaiu O et al (2019) Counter-regulation of regulatory T cells by autoreactive CD8(+) T cells in rheumatoid arthritis. J Autoimmun 99:81–97PubMedCrossRef Cammarata I, Martire C, Citro A, Raimondo D, Fruci D, Melaiu O et al (2019) Counter-regulation of regulatory T cells by autoreactive CD8(+) T cells in rheumatoid arthritis. J Autoimmun 99:81–97PubMedCrossRef
83.
go back to reference Ribon M, Seninet S, Mussard J, Sebbag M, Clavel C, Serre G et al (2019) Neutrophil extracellular traps exert both pro- and anti-inflammatory actions in rheumatoid arthritis that are modulated by C1q and LL-37. J Autoimmun 98:122–131PubMedCrossRef Ribon M, Seninet S, Mussard J, Sebbag M, Clavel C, Serre G et al (2019) Neutrophil extracellular traps exert both pro- and anti-inflammatory actions in rheumatoid arthritis that are modulated by C1q and LL-37. J Autoimmun 98:122–131PubMedCrossRef
84.
go back to reference El Shikh MEM, El Sayed R, Nerviani A, Goldmann K, John CR, Hands R et al (2019) Extracellular traps and PAD4 released by macrophages induce citrullination and auto-antibody production in autoimmune arthritis. J Autoimmun 105:102297PubMedCrossRef El Shikh MEM, El Sayed R, Nerviani A, Goldmann K, John CR, Hands R et al (2019) Extracellular traps and PAD4 released by macrophages induce citrullination and auto-antibody production in autoimmune arthritis. J Autoimmun 105:102297PubMedCrossRef
85.
go back to reference Peene I, Elewaut D (2019) Changing the wolf from outside: how microbiota trigger systemic lupus erythematosus. Ann Rheum Dis 78:867–869PubMedCrossRef Peene I, Elewaut D (2019) Changing the wolf from outside: how microbiota trigger systemic lupus erythematosus. Ann Rheum Dis 78:867–869PubMedCrossRef
86.
go back to reference Silverman GJ, Azzouz DF (2019) Response to: 'The level of peripheral regulatory T cells is linked to changes in gut commensal microflora in patients with systemic lupus erythematosus' by Zhang et al and the phylogeny of a candidate pathobiont in lupus nephritis. Ann Rheum Dis Silverman GJ, Azzouz DF (2019) Response to: 'The level of peripheral regulatory T cells is linked to changes in gut commensal microflora in patients with systemic lupus erythematosus' by Zhang et al and the phylogeny of a candidate pathobiont in lupus nephritis. Ann Rheum Dis
87.
go back to reference van Dijkhuizen EHP, Del Chierico F, Malattia C, Russo A, Pires Marafon D, Ter Haar NM et al (2019) Microbiome analytics of the gut microbiota in patients with juvenile idiopathic arthritis: a longitudinal observational cohort study. Arthritis Rheum 71:1000–1010CrossRef van Dijkhuizen EHP, Del Chierico F, Malattia C, Russo A, Pires Marafon D, Ter Haar NM et al (2019) Microbiome analytics of the gut microbiota in patients with juvenile idiopathic arthritis: a longitudinal observational cohort study. Arthritis Rheum 71:1000–1010CrossRef
88.
go back to reference Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM et al (2019) HLA alleles associated with risk of ankylosing spondylitis and rheumatoid arthritis influence the gut microbiome. Arthritis Rheum 71:1642–1650CrossRef Asquith M, Sternes PR, Costello ME, Karstens L, Diamond S, Martin TM et al (2019) HLA alleles associated with risk of ankylosing spondylitis and rheumatoid arthritis influence the gut microbiome. Arthritis Rheum 71:1642–1650CrossRef
89.
go back to reference Zhou C, Zhao H, Xiao XY, Chen BD, Guo RJ, Wang Q et al (2020) Metagenomic profiling of the pro-inflammatory gut microbiota in ankylosing spondylitis. J Autoimmun 107:102360PubMedCrossRef Zhou C, Zhao H, Xiao XY, Chen BD, Guo RJ, Wang Q et al (2020) Metagenomic profiling of the pro-inflammatory gut microbiota in ankylosing spondylitis. J Autoimmun 107:102360PubMedCrossRef
90.
go back to reference Wells PM, Williams FMK, Matey-Hernandez ML, Menni C, Steves CJ (2019) RA and the microbiome: Do host genetic factors provide the link? J Autoimmun 99:104–115PubMedPubMedCentralCrossRef Wells PM, Williams FMK, Matey-Hernandez ML, Menni C, Steves CJ (2019) RA and the microbiome: Do host genetic factors provide the link? J Autoimmun 99:104–115PubMedPubMedCentralCrossRef
91.
go back to reference Zhao CN, Xu Z, Wu GC, Mao YM, Liu LN, Qian W et al (2019) Emerging role of air pollution in autoimmune diseases. Autoimmun Rev 18:607–614PubMedCrossRef Zhao CN, Xu Z, Wu GC, Mao YM, Liu LN, Qian W et al (2019) Emerging role of air pollution in autoimmune diseases. Autoimmun Rev 18:607–614PubMedCrossRef
92.
go back to reference Yang XK, Liu J, Chen SY, Li M, Zhang MM, Leng RX et al (2019) UBASH3A gene polymorphisms and expression profile in rheumatoid arthritis. Autoimmunity 52:21–26PubMedCrossRef Yang XK, Liu J, Chen SY, Li M, Zhang MM, Leng RX et al (2019) UBASH3A gene polymorphisms and expression profile in rheumatoid arthritis. Autoimmunity 52:21–26PubMedCrossRef
93.
go back to reference Regueiro C, Rodriguez-Rodriguez L, Triguero-Martinez A, Nuno L, Castano-Nunez AL, Villalva A et al (2019) Specific association of HLA-DRB1*03 with anti-carbamylated protein antibodies in patients with rheumatoid arthritis. Arthritis Rheum 71:331–339CrossRef Regueiro C, Rodriguez-Rodriguez L, Triguero-Martinez A, Nuno L, Castano-Nunez AL, Villalva A et al (2019) Specific association of HLA-DRB1*03 with anti-carbamylated protein antibodies in patients with rheumatoid arthritis. Arthritis Rheum 71:331–339CrossRef
94.
go back to reference Plant D, Maciejewski M, Smith S, Nair N, Hyrich K, Ziemek D et al (2019) Profiling of gene expression biomarkers as a classifier of methotrexate nonresponse in patients with rheumatoid arthritis. Arthritis Rheum 71:678–684CrossRef Plant D, Maciejewski M, Smith S, Nair N, Hyrich K, Ziemek D et al (2019) Profiling of gene expression biomarkers as a classifier of methotrexate nonresponse in patients with rheumatoid arthritis. Arthritis Rheum 71:678–684CrossRef
95.
go back to reference Shen F, Verma AH, Volk A, Jones B, Coleman BM, Loza MJ et al (2019) Combined blockade of TNF-alpha and IL-17A alleviates progression of collagen-induced arthritis without causing serious infections in mice. J Immunol 202:2017–2026PubMedCrossRef Shen F, Verma AH, Volk A, Jones B, Coleman BM, Loza MJ et al (2019) Combined blockade of TNF-alpha and IL-17A alleviates progression of collagen-induced arthritis without causing serious infections in mice. J Immunol 202:2017–2026PubMedCrossRef
96.
go back to reference Lorenzetti R, Janowska I, Smulski CR, Frede N, Henneberger N, Walter L et al (2019) Abatacept modulates CD80 and CD86 expression and memory formation in human B-cells. J Autoimmun 101:145–152PubMedCrossRef Lorenzetti R, Janowska I, Smulski CR, Frede N, Henneberger N, Walter L et al (2019) Abatacept modulates CD80 and CD86 expression and memory formation in human B-cells. J Autoimmun 101:145–152PubMedCrossRef
97.
go back to reference De Santis M, Isailovic N, Generali E, Ceribelli A, Altamore L, Real-Fernandez F et al (2019) Humoral response against LL-37 in psoriatic disease: comment on the article by Yuan et al. Arthritis Rheum 71:1964–1965CrossRef De Santis M, Isailovic N, Generali E, Ceribelli A, Altamore L, Real-Fernandez F et al (2019) Humoral response against LL-37 in psoriatic disease: comment on the article by Yuan et al. Arthritis Rheum 71:1964–1965CrossRef
98.
go back to reference Yuan Y, Qiu J, Lin ZT, Li W, Haley C, Mui UN et al (2019) Identification of novel autoantibodies associated with psoriatic arthritis. Arthritis Rheum 71:941–951CrossRef Yuan Y, Qiu J, Lin ZT, Li W, Haley C, Mui UN et al (2019) Identification of novel autoantibodies associated with psoriatic arthritis. Arthritis Rheum 71:941–951CrossRef
99.
go back to reference Sobchak C, Akhtari S, Harvey P, Gladman D, Chandran V, Cook R et al (2019) Value of carotid ultrasound in cardiovascular risk stratification in patients with psoriatic disease. Arthritis Rheum 71:1651–1659CrossRef Sobchak C, Akhtari S, Harvey P, Gladman D, Chandran V, Cook R et al (2019) Value of carotid ultrasound in cardiovascular risk stratification in patients with psoriatic disease. Arthritis Rheum 71:1651–1659CrossRef
100.
go back to reference Savage L, Goodfield M, Horton L, Watad A, Hensor E, Emery P et al (2019) Regression of peripheral subclinical enthesopathy in therapy-naive patients treated with ustekinumab for moderate-to-severe chronic plaque psoriasis: a fifty-two-week, prospective, open-label feasibility study. Arthritis Rheum 71:626–631CrossRef Savage L, Goodfield M, Horton L, Watad A, Hensor E, Emery P et al (2019) Regression of peripheral subclinical enthesopathy in therapy-naive patients treated with ustekinumab for moderate-to-severe chronic plaque psoriasis: a fifty-two-week, prospective, open-label feasibility study. Arthritis Rheum 71:626–631CrossRef
101.
go back to reference Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A et al (2019) Special article: 2018 American College of Rheumatology/National Psoriasis Foundation guideline for the treatment of psoriatic arthritis. Arthritis Rheum 71:5–32CrossRef Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A et al (2019) Special article: 2018 American College of Rheumatology/National Psoriasis Foundation guideline for the treatment of psoriatic arthritis. Arthritis Rheum 71:5–32CrossRef
102.
go back to reference Chattopadhyay A, Sharma SK, Naidu S, Dhir V (2019) Failure of etanercept and methotrexate combination therapy to surpass etanercept monotherapy in psoriatic arthritis-What about the joint counts? Comment on the Article by Mease et al. Arthritis Rheum 71:1965–1966CrossRef Chattopadhyay A, Sharma SK, Naidu S, Dhir V (2019) Failure of etanercept and methotrexate combination therapy to surpass etanercept monotherapy in psoriatic arthritis-What about the joint counts? Comment on the Article by Mease et al. Arthritis Rheum 71:1965–1966CrossRef
103.
go back to reference Siebert S, Sweet K, Dasgupta B, Campbell K, McInnes IB, Loza MJ (2019) Responsiveness of serum C-reactive protein, interleukin-17A, and interleukin-17F levels to ustekinumab in psoriatic arthritis: lessons from two phase III, multicenter, double-blind, placebo-controlled trials. Arthritis Rheum 71:1660–1669CrossRef Siebert S, Sweet K, Dasgupta B, Campbell K, McInnes IB, Loza MJ (2019) Responsiveness of serum C-reactive protein, interleukin-17A, and interleukin-17F levels to ustekinumab in psoriatic arthritis: lessons from two phase III, multicenter, double-blind, placebo-controlled trials. Arthritis Rheum 71:1660–1669CrossRef
104.
go back to reference Merola JF, Ogdie A (2019) SEAM-PsA: seems like methotrexate works in psoriatic arthritis? Arthritis Rheum 71:1027–1029CrossRef Merola JF, Ogdie A (2019) SEAM-PsA: seems like methotrexate works in psoriatic arthritis? Arthritis Rheum 71:1027–1029CrossRef
105.
go back to reference Mease PJ, Gladman DD, Collier DH, Ritchlin CT, Helliwell PS, Liu L et al (2019) Etanercept and methotrexate as monotherapy or in combination for psoriatic arthritis: primary results from a randomized, controlled phase III trial. Arthritis Rheum 71:1112–1124CrossRef Mease PJ, Gladman DD, Collier DH, Ritchlin CT, Helliwell PS, Liu L et al (2019) Etanercept and methotrexate as monotherapy or in combination for psoriatic arthritis: primary results from a randomized, controlled phase III trial. Arthritis Rheum 71:1112–1124CrossRef
106.
go back to reference Agrawal M, Shah S, Patel A, Pinotti R, Colombel JF, Burisch J (2019) Changing epidemiology of immune-mediated inflammatory diseases in immigrants: a systematic review of population-based studies. J Autoimmun 105:102303PubMedCrossRefPubMedCentral Agrawal M, Shah S, Patel A, Pinotti R, Colombel JF, Burisch J (2019) Changing epidemiology of immune-mediated inflammatory diseases in immigrants: a systematic review of population-based studies. J Autoimmun 105:102303PubMedCrossRefPubMedCentral
107.
go back to reference Bernink JH, Ohne Y, Teunissen MBM, Wang J, Wu J, Krabbendam L et al (2019) c-Kit-positive ILC2s exhibit an ILC3-like signature that may contribute to IL-17-mediated pathologies. Nat Immunol 20:992–1003PubMedCrossRef Bernink JH, Ohne Y, Teunissen MBM, Wang J, Wu J, Krabbendam L et al (2019) c-Kit-positive ILC2s exhibit an ILC3-like signature that may contribute to IL-17-mediated pathologies. Nat Immunol 20:992–1003PubMedCrossRef
108.
go back to reference Chimenti MS, Caso F, Alivernini S, De Martino E, Costa L, Tolusso B et al (2019) Amplifying the concept of psoriatic arthritis: the role of autoimmunity in systemic psoriatic disease. Autoimmun Rev 18:565–575PubMedCrossRef Chimenti MS, Caso F, Alivernini S, De Martino E, Costa L, Tolusso B et al (2019) Amplifying the concept of psoriatic arthritis: the role of autoimmunity in systemic psoriatic disease. Autoimmun Rev 18:565–575PubMedCrossRef
109.
go back to reference Bergua C, Chiavelli H, Allenbach Y, Arouche-Delaperche L, Arnoult C, Bourdenet G et al (2019) In vivo pathogenicity of IgG from patients with anti-SRP or anti-HMGCR autoantibodies in immune-mediated necrotising myopathy. Ann Rheum Dis 78:131–139PubMedCrossRef Bergua C, Chiavelli H, Allenbach Y, Arouche-Delaperche L, Arnoult C, Bourdenet G et al (2019) In vivo pathogenicity of IgG from patients with anti-SRP or anti-HMGCR autoantibodies in immune-mediated necrotising myopathy. Ann Rheum Dis 78:131–139PubMedCrossRef
110.
go back to reference Betancur JF, Gomez-Puerta JA (2020) Antinuclear antibodies mitotic patterns and their clinical associations. Ann Rheum Dis 79:e63PubMedCrossRef Betancur JF, Gomez-Puerta JA (2020) Antinuclear antibodies mitotic patterns and their clinical associations. Ann Rheum Dis 79:e63PubMedCrossRef
111.
go back to reference Damoiseaux J, Andrade LEC, Carballo OG, Conrad K, Francescantonio PLC, Fritzler MJ et al (2019) Clinical relevance of HEp-2 indirect immunofluorescent patterns: the International Consensus on ANA patterns (ICAP) perspective. Ann Rheum Dis 78:879–889PubMedCrossRef Damoiseaux J, Andrade LEC, Carballo OG, Conrad K, Francescantonio PLC, Fritzler MJ et al (2019) Clinical relevance of HEp-2 indirect immunofluorescent patterns: the International Consensus on ANA patterns (ICAP) perspective. Ann Rheum Dis 78:879–889PubMedCrossRef
112.
go back to reference Deng C, Qu X, Cheng S, Zeng X, Li Y, Fei Y (2019) Decision-making value of nuclear dense fine speckled pattern in systemic autoimmune rheumatic disease: trick or treat? Ann Rheum Dis Deng C, Qu X, Cheng S, Zeng X, Li Y, Fei Y (2019) Decision-making value of nuclear dense fine speckled pattern in systemic autoimmune rheumatic disease: trick or treat? Ann Rheum Dis
113.
go back to reference Greco M, Garcia de Yebenes MJ, Alarcon I, Brandy-Garcia AM, Rua-Figueroa I, Loza E et al (2020) Antisynthetase antibodies in clinical laboratories: the importance of clinical correlation and indirect immunofluorescence. Response to: Comment on: 'Idiopathic inflammatory myopathies and antisynthetase syndrome: contribution of antisynthetase antibodies to improve current classification criteria' by Greco et al' by Knitza et al. Ann Rheum Dis 79:e87PubMedCrossRef Greco M, Garcia de Yebenes MJ, Alarcon I, Brandy-Garcia AM, Rua-Figueroa I, Loza E et al (2020) Antisynthetase antibodies in clinical laboratories: the importance of clinical correlation and indirect immunofluorescence. Response to: Comment on: 'Idiopathic inflammatory myopathies and antisynthetase syndrome: contribution of antisynthetase antibodies to improve current classification criteria' by Greco et al' by Knitza et al. Ann Rheum Dis 79:e87PubMedCrossRef
114.
go back to reference Lee AYS, Beroukas D, Roberts-Thomson PJ (2020) Utility of the HEp-2000 antinuclear antibody substrate. Ann Rheum Dis 79:e67PubMedCrossRef Lee AYS, Beroukas D, Roberts-Thomson PJ (2020) Utility of the HEp-2000 antinuclear antibody substrate. Ann Rheum Dis 79:e67PubMedCrossRef
115.
go back to reference Rothwell S, Chinoy H, Lamb JA, Miller FW, Rider LG, Wedderburn LR et al (2019) Focused HLA analysis in Caucasians with myositis identifies significant associations with autoantibody subgroups. Ann Rheum Dis 78:996–1002PubMedCrossRef Rothwell S, Chinoy H, Lamb JA, Miller FW, Rider LG, Wedderburn LR et al (2019) Focused HLA analysis in Caucasians with myositis identifies significant associations with autoantibody subgroups. Ann Rheum Dis 78:996–1002PubMedCrossRef
116.
go back to reference Sabbagh S, Pinal-Fernandez I, Kishi T, Targoff IN, Miller FW, Rider LG et al (2019) Anti-Ro52 autoantibodies are associated with interstitial lung disease and more severe disease in patients with juvenile myositis. Ann Rheum Dis 78:988–995PubMedCrossRef Sabbagh S, Pinal-Fernandez I, Kishi T, Targoff IN, Miller FW, Rider LG et al (2019) Anti-Ro52 autoantibodies are associated with interstitial lung disease and more severe disease in patients with juvenile myositis. Ann Rheum Dis 78:988–995PubMedCrossRef
117.
go back to reference Spielmann L, Nespola B, Severac F, Andres E, Kessler R, Guffroy A et al (2019) Anti-Ku syndrome with elevated CK and anti-Ku syndrome with anti-dsDNA are two distinct entities with different outcomes. Ann Rheum Dis 78:1101–1106PubMedCrossRef Spielmann L, Nespola B, Severac F, Andres E, Kessler R, Guffroy A et al (2019) Anti-Ku syndrome with elevated CK and anti-Ku syndrome with anti-dsDNA are two distinct entities with different outcomes. Ann Rheum Dis 78:1101–1106PubMedCrossRef
118.
go back to reference van Delft MAM, van der Woude D, Toes REM, Trouw LA (2019) Secretory form of rheumatoid arthritis-associated autoantibodies in serum are mainly of the IgM isotype, suggesting a continuous reactivation of autoantibody responses at mucosal surfaces. Ann Rheum Dis 78:146–148PubMedCrossRef van Delft MAM, van der Woude D, Toes REM, Trouw LA (2019) Secretory form of rheumatoid arthritis-associated autoantibodies in serum are mainly of the IgM isotype, suggesting a continuous reactivation of autoantibody responses at mucosal surfaces. Ann Rheum Dis 78:146–148PubMedCrossRef
119.
go back to reference Vulsteke JB, Van Hoovels L, Willems P, Vander Cruyssen B, Vanderschueren S, Westhovens R et al. (2019) Titre-specific positive predictive value of antinuclear antibody patterns. Ann Rheum Dis Vulsteke JB, Van Hoovels L, Willems P, Vander Cruyssen B, Vanderschueren S, Westhovens R et al. (2019) Titre-specific positive predictive value of antinuclear antibody patterns. Ann Rheum Dis
120.
go back to reference Andrianova IA, Ponomareva AA, Mordakhanova ER, Le Minh G, Daminova AG, Nevzorova TA et al (2020) In systemic lupus erythematosus anti-dsDNA antibodies can promote thrombosis through direct platelet activation. J Autoimmun 107:102355PubMedCrossRef Andrianova IA, Ponomareva AA, Mordakhanova ER, Le Minh G, Daminova AG, Nevzorova TA et al (2020) In systemic lupus erythematosus anti-dsDNA antibodies can promote thrombosis through direct platelet activation. J Autoimmun 107:102355PubMedCrossRef
121.
go back to reference Germar K, Fehres CM, Scherer HU, van Uden N, Pollastro S, Yeremenko N et al (2019) Generation and characterization of anti-citrullinated protein antibody-producing B cell clones from rheumatoid arthritis patients. Arthritis Rheum 71:340–350CrossRef Germar K, Fehres CM, Scherer HU, van Uden N, Pollastro S, Yeremenko N et al (2019) Generation and characterization of anti-citrullinated protein antibody-producing B cell clones from rheumatoid arthritis patients. Arthritis Rheum 71:340–350CrossRef
122.
go back to reference Hoshino-Negishi K, Ohkuro M, Nakatani T, Kuboi Y, Nishimura M, Ida Y et al (2019) Role of anti-fractalkine antibody in suppression of joint destruction by inhibiting migration of osteoclast precursors to the synovium in experimental arthritis. Arthritis Rheum 71:222–231CrossRef Hoshino-Negishi K, Ohkuro M, Nakatani T, Kuboi Y, Nishimura M, Ida Y et al (2019) Role of anti-fractalkine antibody in suppression of joint destruction by inhibiting migration of osteoclast precursors to the synovium in experimental arthritis. Arthritis Rheum 71:222–231CrossRef
123.
go back to reference Kelmenson LB, Wagner BD, McNair BK, Frazer-Abel A, Demoruelle MK, Bergstedt DT et al (2020) Timing of elevations of autoantibody isotypes prior to diagnosis of rheumatoid arthritis. Arthritis Rheum 72:251–261CrossRef Kelmenson LB, Wagner BD, McNair BK, Frazer-Abel A, Demoruelle MK, Bergstedt DT et al (2020) Timing of elevations of autoantibody isotypes prior to diagnosis of rheumatoid arthritis. Arthritis Rheum 72:251–261CrossRef
124.
go back to reference Kongpachith S, Lingampalli N, Ju CH, Blum LK, Lu DR, Elliott SE et al (2019) Affinity maturation of the anti-citrullinated protein antibody paratope drives epitope spreading and polyreactivity in rheumatoid arthritis. Arthritis Rheum 71:507–517CrossRef Kongpachith S, Lingampalli N, Ju CH, Blum LK, Lu DR, Elliott SE et al (2019) Affinity maturation of the anti-citrullinated protein antibody paratope drives epitope spreading and polyreactivity in rheumatoid arthritis. Arthritis Rheum 71:507–517CrossRef
125.
go back to reference Lo KC, Sullivan E, Bannen RM, Jin H, Rowe M, Li H et al (2020) Comprehensive profiling of the rheumatoid arthritis antibody repertoire. Arthritis Rheum 72:242–250CrossRef Lo KC, Sullivan E, Bannen RM, Jin H, Rowe M, Li H et al (2020) Comprehensive profiling of the rheumatoid arthritis antibody repertoire. Arthritis Rheum 72:242–250CrossRef
126.
go back to reference Schwenzer A, Quirke AM, Montgomery AB, Venables PJ, Sayles HR, Schlumberger W et al (2019) Time to include fine specificity anti-citrullinated protein antibodies in the routine diagnosis and management of rheumatoid arthritis? Arthritis Rheum 71:476–478CrossRef Schwenzer A, Quirke AM, Montgomery AB, Venables PJ, Sayles HR, Schlumberger W et al (2019) Time to include fine specificity anti-citrullinated protein antibodies in the routine diagnosis and management of rheumatoid arthritis? Arthritis Rheum 71:476–478CrossRef
127.
go back to reference Shelef MA (2019) New relationships for old autoantibodies in rheumatoid arthritis. Arthritis Rheum 71:1396–1399CrossRef Shelef MA (2019) New relationships for old autoantibodies in rheumatoid arthritis. Arthritis Rheum 71:1396–1399CrossRef
128.
go back to reference Zheng Z, Mergaert AM, Fahmy LM, Bawadekar M, Holmes CL, Ong IM et al (2020) Disordered antigens and epitope overlap between anti-citrullinated protein antibodies and rheumatoid factor in rheumatoid arthritis. Arthritis Rheum 72:262–272CrossRef Zheng Z, Mergaert AM, Fahmy LM, Bawadekar M, Holmes CL, Ong IM et al (2020) Disordered antigens and epitope overlap between anti-citrullinated protein antibodies and rheumatoid factor in rheumatoid arthritis. Arthritis Rheum 72:262–272CrossRef
129.
go back to reference Ge C, Xu B, Liang B, Lonnblom E, Lundstrom SL, Zubarev RA et al (2019) Structural basis of cross-reactivity of anti-citrullinated protein antibodies. Arthritis Rheum 71:210–221CrossRef Ge C, Xu B, Liang B, Lonnblom E, Lundstrom SL, Zubarev RA et al (2019) Structural basis of cross-reactivity of anti-citrullinated protein antibodies. Arthritis Rheum 71:210–221CrossRef
130.
go back to reference Hafkenscheid L, de Moel E, Smolik I, Tanner S, Meng X, Jansen BC et al (2019) N-linked glycans in the variable domain of IgG anti-citrullinated protein antibodies predict the development of rheumatoid arthritis. Arthritis Rheum 71:1626–1633CrossRef Hafkenscheid L, de Moel E, Smolik I, Tanner S, Meng X, Jansen BC et al (2019) N-linked glycans in the variable domain of IgG anti-citrullinated protein antibodies predict the development of rheumatoid arthritis. Arthritis Rheum 71:1626–1633CrossRef
131.
go back to reference Holmdahl R (2019) The specificity of monoclonal anti-citrullinated protein antibodies: comment on the article by Steen et al. Arthritis Rheum 71:324–325CrossRef Holmdahl R (2019) The specificity of monoclonal anti-citrullinated protein antibodies: comment on the article by Steen et al. Arthritis Rheum 71:324–325CrossRef
132.
go back to reference Jain A, Shah H, Simonsick EM, Metter EJ, Mangold L, Humphreys E et al (2019) Angiotensin receptor autoantibodies as exposures that modify disease progression: cross sectional, longitudinal and in vitro studies of prostate cancer. J Transl Autoimmun 2 Jain A, Shah H, Simonsick EM, Metter EJ, Mangold L, Humphreys E et al (2019) Angiotensin receptor autoantibodies as exposures that modify disease progression: cross sectional, longitudinal and in vitro studies of prostate cancer. J Transl Autoimmun 2
133.
go back to reference Stathopoulos P, Chastre A, Waters P, Irani S, Fichtner ML, Benotti ES et al (2019) Autoantibodies against neurologic antigens in nonneurologic autoimmunity. J Immunol 202:2210–2219PubMedPubMedCentralCrossRef Stathopoulos P, Chastre A, Waters P, Irani S, Fichtner ML, Benotti ES et al (2019) Autoantibodies against neurologic antigens in nonneurologic autoimmunity. J Immunol 202:2210–2219PubMedPubMedCentralCrossRef
134.
go back to reference Fresquet M, Rhoden SJ, Jowitt TA, McKenzie EA, Roberts I, Lennon R et al (2020) Autoantigens PLA2R and THSD7A in membranous nephropathy share a common epitope motif in the N-terminal domain. J Autoimmun 106:102308PubMedPubMedCentralCrossRef Fresquet M, Rhoden SJ, Jowitt TA, McKenzie EA, Roberts I, Lennon R et al (2020) Autoantigens PLA2R and THSD7A in membranous nephropathy share a common epitope motif in the N-terminal domain. J Autoimmun 106:102308PubMedPubMedCentralCrossRef
135.
go back to reference Alhomaidan HT, Rasheed N, Almatrafi S, Al-Rashdi FH, Rasheed Z (2019) Bisphenol A modified DNA: a possible immunogenic stimulus for anti-DNA autoantibodies in systemic lupus erythematosus. Autoimmunity 52:272–280PubMedCrossRef Alhomaidan HT, Rasheed N, Almatrafi S, Al-Rashdi FH, Rasheed Z (2019) Bisphenol A modified DNA: a possible immunogenic stimulus for anti-DNA autoantibodies in systemic lupus erythematosus. Autoimmunity 52:272–280PubMedCrossRef
136.
go back to reference Acosta-Herrera M, Kerick M, Gonzalez-Serna D, Wijmenga C, Franke A, Gregersen PK et al (2019) Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases. Ann Rheum Dis 78:311–319PubMedCrossRef Acosta-Herrera M, Kerick M, Gonzalez-Serna D, Wijmenga C, Franke A, Gregersen PK et al (2019) Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases. Ann Rheum Dis 78:311–319PubMedCrossRef
137.
go back to reference Akizuki S, Ishigaki K, Kochi Y, Law SM, Matsuo K, Ohmura K et al (2019) PLD4 is a genetic determinant to systemic lupus erythematosus and involved in murine autoimmune phenotypes. Ann Rheum Dis 78:509–518PubMedCrossRef Akizuki S, Ishigaki K, Kochi Y, Law SM, Matsuo K, Ohmura K et al (2019) PLD4 is a genetic determinant to systemic lupus erythematosus and involved in murine autoimmune phenotypes. Ann Rheum Dis 78:509–518PubMedCrossRef
138.
go back to reference Tkachenko O, Lapin S, Maslyansky A, Myachikova V, Guseva V, Belolipetskaia E et al (2019) Influence of HLA-DRB1 susceptibility alleles on the autoantibodies spectrum of systemic lupus erythematosus in European part of Russia. Autoimmun Rev 18:558–560PubMedCrossRef Tkachenko O, Lapin S, Maslyansky A, Myachikova V, Guseva V, Belolipetskaia E et al (2019) Influence of HLA-DRB1 susceptibility alleles on the autoantibodies spectrum of systemic lupus erythematosus in European part of Russia. Autoimmun Rev 18:558–560PubMedCrossRef
139.
go back to reference Shah AA, Laiho M, Rosen A, Casciola-Rosen L (2019) Protective effect against cancer of antibodies to the large subunits of both RNA polymerases I and III in scleroderma. Arthritis Rheum 71:1571–1579CrossRef Shah AA, Laiho M, Rosen A, Casciola-Rosen L (2019) Protective effect against cancer of antibodies to the large subunits of both RNA polymerases I and III in scleroderma. Arthritis Rheum 71:1571–1579CrossRef
140.
go back to reference Pinal-Fernandez I, Amici DR, Parks CA, Derfoul A, Casal-Dominguez M, Pak K et al (2019) Myositis autoantigen expression correlates with muscle regeneration but not autoantibody specificity. Arthritis Rheum 71:1371–1376CrossRef Pinal-Fernandez I, Amici DR, Parks CA, Derfoul A, Casal-Dominguez M, Pak K et al (2019) Myositis autoantigen expression correlates with muscle regeneration but not autoantibody specificity. Arthritis Rheum 71:1371–1376CrossRef
141.
go back to reference Mosanya CH, Isaacs JD (2019) Tolerising cellular therapies: what is their promise for autoimmune disease? Ann Rheum Dis 78:297–310PubMedCrossRef Mosanya CH, Isaacs JD (2019) Tolerising cellular therapies: what is their promise for autoimmune disease? Ann Rheum Dis 78:297–310PubMedCrossRef
142.
go back to reference Scheinberg M, Azevedo V (2019) The future landscape of biosimilars in rheumatology: Where we are where we are going. Autoimmun Rev 18:203–208PubMedCrossRef Scheinberg M, Azevedo V (2019) The future landscape of biosimilars in rheumatology: Where we are where we are going. Autoimmun Rev 18:203–208PubMedCrossRef
143.
go back to reference Muhammad Yusoff F, Wong KK, Mohd Redzwan N (2020) Th1, Th2, and Th17 cytokines in systemic lupus erythematosus. Autoimmunity 53:8–20PubMedCrossRef Muhammad Yusoff F, Wong KK, Mohd Redzwan N (2020) Th1, Th2, and Th17 cytokines in systemic lupus erythematosus. Autoimmunity 53:8–20PubMedCrossRef
144.
go back to reference Rahman S, Sagar D, Hanna RN, Lightfoot YL, Mistry P, Smith CK et al (2019) Low-density granulocytes activate T cells and demonstrate a non-suppressive role in systemic lupus erythematosus. Ann Rheum Dis 78:957–966PubMedCrossRef Rahman S, Sagar D, Hanna RN, Lightfoot YL, Mistry P, Smith CK et al (2019) Low-density granulocytes activate T cells and demonstrate a non-suppressive role in systemic lupus erythematosus. Ann Rheum Dis 78:957–966PubMedCrossRef
145.
go back to reference Chan KK, Bass AR (2019) Checkpoint inhibitor-induced polymyalgia rheumatica controlled by cobimetinib, a MEK 1/2 inhibitor. Ann Rheum Dis 78:e70PubMedCrossRef Chan KK, Bass AR (2019) Checkpoint inhibitor-induced polymyalgia rheumatica controlled by cobimetinib, a MEK 1/2 inhibitor. Ann Rheum Dis 78:e70PubMedCrossRef
146.
go back to reference Delyon J, Brunet-Possenti F, Leonard-Louis S, Arangalage D, Baudet M, Baroudjian B et al (2019) Immune checkpoint inhibitor rechallenge in patients with immune-related myositis. Ann Rheum Dis 78:e129PubMedCrossRef Delyon J, Brunet-Possenti F, Leonard-Louis S, Arangalage D, Baudet M, Baroudjian B et al (2019) Immune checkpoint inhibitor rechallenge in patients with immune-related myositis. Ann Rheum Dis 78:e129PubMedCrossRef
147.
go back to reference Kostine M, Cappelli LC, Calabrese C, Calabrese LH, Bingham CO 3rd, Richez C et al (2019) Addressing immune-related adverse events of cancer immunotherapy: how prepared are rheumatologists? Ann Rheum Dis 78:860–862PubMedCrossRef Kostine M, Cappelli LC, Calabrese C, Calabrese LH, Bingham CO 3rd, Richez C et al (2019) Addressing immune-related adverse events of cancer immunotherapy: how prepared are rheumatologists? Ann Rheum Dis 78:860–862PubMedCrossRef
148.
go back to reference Mammen AL, Rajan A, Pak K, Lehky T, Casciola-Rosen L, Donahue RN et al (2019) Pre-existing antiacetylcholine receptor autoantibodies and B cell lymphopaenia are associated with the development of myositis in patients with thymoma treated with avelumab, an immune checkpoint inhibitor targeting programmed death-ligand 1. Ann Rheum Dis 78:150–152PubMedCrossRef Mammen AL, Rajan A, Pak K, Lehky T, Casciola-Rosen L, Donahue RN et al (2019) Pre-existing antiacetylcholine receptor autoantibodies and B cell lymphopaenia are associated with the development of myositis in patients with thymoma treated with avelumab, an immune checkpoint inhibitor targeting programmed death-ligand 1. Ann Rheum Dis 78:150–152PubMedCrossRef
149.
go back to reference Michot JM, Fusellier M, Champiat S, Velter C, Baldini C, Voisin AL et al (2019) Drug-induced lupus erythematosus following immunotherapy with anti-programmed death-(ligand) 1. Ann Rheum Dis 78:e67PubMedCrossRef Michot JM, Fusellier M, Champiat S, Velter C, Baldini C, Voisin AL et al (2019) Drug-induced lupus erythematosus following immunotherapy with anti-programmed death-(ligand) 1. Ann Rheum Dis 78:e67PubMedCrossRef
150.
go back to reference Schaeverbeke T, Kostine M (2019) Response to: 'Checkpoint inhibitors and arthritis: seeking balance between victories and defeats' by Moura and Moura. Ann Rheum Dis 78:e92PubMedCrossRef Schaeverbeke T, Kostine M (2019) Response to: 'Checkpoint inhibitors and arthritis: seeking balance between victories and defeats' by Moura and Moura. Ann Rheum Dis 78:e92PubMedCrossRef
151.
go back to reference McGonagle D, Bragazzi NL, Amital H, Watad A (2020) Mechanistic classification of immune checkpoint inhibitor toxicity as a pointer to minimal treatment strategies to further improve survival. Autoimmun Rev 19:102456PubMedCrossRef McGonagle D, Bragazzi NL, Amital H, Watad A (2020) Mechanistic classification of immune checkpoint inhibitor toxicity as a pointer to minimal treatment strategies to further improve survival. Autoimmun Rev 19:102456PubMedCrossRef
152.
go back to reference Kulik L, Laskowski J, Renner B, Woolaver R, Zhang L, Lyubchenko T et al (2019) Targeting the immune complex-bound complement C3d ligand as a novel therapy for lupus. J Immunol 203:3136–3147PubMedPubMedCentralCrossRef Kulik L, Laskowski J, Renner B, Woolaver R, Zhang L, Lyubchenko T et al (2019) Targeting the immune complex-bound complement C3d ligand as a novel therapy for lupus. J Immunol 203:3136–3147PubMedPubMedCentralCrossRef
153.
go back to reference Khan H, Sureda A, Belwal T, Cetinkaya S, Suntar I, Tejada S et al (2019) Polyphenols in the treatment of autoimmune diseases. Autoimmun Rev 18:647–657PubMedPubMedCentralCrossRef Khan H, Sureda A, Belwal T, Cetinkaya S, Suntar I, Tejada S et al (2019) Polyphenols in the treatment of autoimmune diseases. Autoimmun Rev 18:647–657PubMedPubMedCentralCrossRef
154.
go back to reference Kim SY, Yu M, Morin EE, Kang J, Kaplan MJ, Schwendeman A (2020) High-density lipoprotein in lupus: disease biomarkers and potential therapeutic strategy. Arthritis Rheum 72:20–30CrossRef Kim SY, Yu M, Morin EE, Kang J, Kaplan MJ, Schwendeman A (2020) High-density lipoprotein in lupus: disease biomarkers and potential therapeutic strategy. Arthritis Rheum 72:20–30CrossRef
155.
go back to reference Kitas GD, Nightingale P, Armitage J, Sattar N, Belch JJF, Symmons DPM (2019) A multicenter, randomized, placebo-controlled trial of atorvastatin for the primary prevention of cardiovascular events in patients with rheumatoid arthritis. Arthritis Rheum 71:1437–1449CrossRef Kitas GD, Nightingale P, Armitage J, Sattar N, Belch JJF, Symmons DPM (2019) A multicenter, randomized, placebo-controlled trial of atorvastatin for the primary prevention of cardiovascular events in patients with rheumatoid arthritis. Arthritis Rheum 71:1437–1449CrossRef
156.
go back to reference Liao KP, Solomon DH (2019) Lipids and cardiovascular risk through the lens of rheumatoid arthritis. Arthritis Rheum 71:1393–1395CrossRef Liao KP, Solomon DH (2019) Lipids and cardiovascular risk through the lens of rheumatoid arthritis. Arthritis Rheum 71:1393–1395CrossRef
157.
go back to reference Lopez-Mejias R, Carmona FD, Genre F, Remuzgo-Martinez S, Gonzalez-Juanatey C, Corrales A et al (2019) Identification of a 3'-untranslated genetic variant of RARB associated with carotid intima-media thickness in rheumatoid arthritis: a genome-wide association study. Arthritis Rheum 71:351–360CrossRef Lopez-Mejias R, Carmona FD, Genre F, Remuzgo-Martinez S, Gonzalez-Juanatey C, Corrales A et al (2019) Identification of a 3'-untranslated genetic variant of RARB associated with carotid intima-media thickness in rheumatoid arthritis: a genome-wide association study. Arthritis Rheum 71:351–360CrossRef
158.
go back to reference Pellicciotta M, Rigoni R, Falcone EL, Holland SM, Villa A, Cassani B (2019) The microbiome and immunodeficiencies: lessons from rare diseases. J Autoimmun 98:132–148PubMedCrossRef Pellicciotta M, Rigoni R, Falcone EL, Holland SM, Villa A, Cassani B (2019) The microbiome and immunodeficiencies: lessons from rare diseases. J Autoimmun 98:132–148PubMedCrossRef
159.
go back to reference Croci S, Bonacini M, Muratore F, Caruso A, Fontana A, Boiardi L et al (2019) The therapeutic potential of tuftsin-phosphorylcholine in giant cell arteritis. J Autoimmun 98:113–121PubMedCrossRef Croci S, Bonacini M, Muratore F, Caruso A, Fontana A, Boiardi L et al (2019) The therapeutic potential of tuftsin-phosphorylcholine in giant cell arteritis. J Autoimmun 98:113–121PubMedCrossRef
160.
go back to reference Tsai HC, Nguyen K, Hashemi E, Engleman E, Hla T, Han MH (2019) Myeloid sphingosine-1-phosphate receptor 1 is important for CNS autoimmunity and neuroinflammation. J Autoimmun 105:102290PubMedCrossRef Tsai HC, Nguyen K, Hashemi E, Engleman E, Hla T, Han MH (2019) Myeloid sphingosine-1-phosphate receptor 1 is important for CNS autoimmunity and neuroinflammation. J Autoimmun 105:102290PubMedCrossRef
161.
go back to reference Alexopoulos H, Dalakas MC (2019) The immunobiology of autoimmune encephalitides. J Autoimmun 104:102339PubMedCrossRef Alexopoulos H, Dalakas MC (2019) The immunobiology of autoimmune encephalitides. J Autoimmun 104:102339PubMedCrossRef
162.
go back to reference Bronge M, Ruhrmann S, Carvalho-Queiroz C, Nilsson OB, Kaiser A, Holmgren E et al (2019) Myelin oligodendrocyte glycoprotein revisited-sensitive detection of MOG-specific T-cells in multiple sclerosis. J Autoimmun 102:38–49PubMedCrossRef Bronge M, Ruhrmann S, Carvalho-Queiroz C, Nilsson OB, Kaiser A, Holmgren E et al (2019) Myelin oligodendrocyte glycoprotein revisited-sensitive detection of MOG-specific T-cells in multiple sclerosis. J Autoimmun 102:38–49PubMedCrossRef
163.
go back to reference Mattapallil MJ, Kielczewski JL, Zarate-Blades CR, St Leger AJ, Raychaudhuri K, Silver PB et al (2019) Interleukin 22 ameliorates neuropathology and protects from central nervous system autoimmunity. J Autoimmun 102:65–76PubMedPubMedCentralCrossRef Mattapallil MJ, Kielczewski JL, Zarate-Blades CR, St Leger AJ, Raychaudhuri K, Silver PB et al (2019) Interleukin 22 ameliorates neuropathology and protects from central nervous system autoimmunity. J Autoimmun 102:65–76PubMedPubMedCentralCrossRef
164.
go back to reference Brate AA, Boyden AW, Itani FR, Pewe LL, Harty JT, Karandikar NJ (2019) Therapeutic intervention in relapsing autoimmune demyelinating disease through induction of myelin-specific regulatory CD8 T cell responses. J Transl Autoimmun 2 Brate AA, Boyden AW, Itani FR, Pewe LL, Harty JT, Karandikar NJ (2019) Therapeutic intervention in relapsing autoimmune demyelinating disease through induction of myelin-specific regulatory CD8 T cell responses. J Transl Autoimmun 2
165.
go back to reference Shemer A, Willis R, Gonzalez EB, Romay-Penabad Z, Shovman O, Shoenfeld Y et al (2019) Oral administration of Domain-I of beta-2glycoprotein-I induces immunological tolerance in experimental murine antiphospholipid syndrome. J Autoimmun 99:98–103PubMedCrossRef Shemer A, Willis R, Gonzalez EB, Romay-Penabad Z, Shovman O, Shoenfeld Y et al (2019) Oral administration of Domain-I of beta-2glycoprotein-I induces immunological tolerance in experimental murine antiphospholipid syndrome. J Autoimmun 99:98–103PubMedCrossRef
166.
go back to reference Qureshi MS, Alsughayyir J, Chhabra M, Ali JM, Goddard MJ, Devine CA et al (2019) Germinal center humoral autoimmunity independently mediates progression of allograft vasculopathy. J Autoimmun 98:44–58PubMedCrossRef Qureshi MS, Alsughayyir J, Chhabra M, Ali JM, Goddard MJ, Devine CA et al (2019) Germinal center humoral autoimmunity independently mediates progression of allograft vasculopathy. J Autoimmun 98:44–58PubMedCrossRef
167.
go back to reference Alvarez-Sierra D, Marin-Sanchez A, Ruiz-Blazquez P, de Jesus GC, Iglesias-Felip C, Gonzalez O et al (2019) Analysis of the PD-1/PD-L1 axis in human autoimmune thyroid disease: insights into pathogenesis and clues to immunotherapy associated thyroid autoimmunity. J Autoimmun 103:102285PubMedCrossRef Alvarez-Sierra D, Marin-Sanchez A, Ruiz-Blazquez P, de Jesus GC, Iglesias-Felip C, Gonzalez O et al (2019) Analysis of the PD-1/PD-L1 axis in human autoimmune thyroid disease: insights into pathogenesis and clues to immunotherapy associated thyroid autoimmunity. J Autoimmun 103:102285PubMedCrossRef
168.
go back to reference Das T, Bergen IM, Koudstaal T, van Hulst JAC, van Loo G, Boonstra A et al (2019) DNGR1-mediated deletion of A20/Tnfaip3 in dendritic cells alters T and B-cell homeostasis and promotes autoimmune liver pathology. J Autoimmun 102:167–178PubMedCrossRef Das T, Bergen IM, Koudstaal T, van Hulst JAC, van Loo G, Boonstra A et al (2019) DNGR1-mediated deletion of A20/Tnfaip3 in dendritic cells alters T and B-cell homeostasis and promotes autoimmune liver pathology. J Autoimmun 102:167–178PubMedCrossRef
169.
go back to reference Terziroli Beretta-Piccoli B, Di Bartolomeo C, Deleonardi G, Grondona AG, Silvestri T, Tesei C et al (2019) Autoimmune liver serology before and after successful treatment of chronic hepatitis C by direct acting antiviral agents. J Autoimmun 102:89–95PubMedCrossRef Terziroli Beretta-Piccoli B, Di Bartolomeo C, Deleonardi G, Grondona AG, Silvestri T, Tesei C et al (2019) Autoimmune liver serology before and after successful treatment of chronic hepatitis C by direct acting antiviral agents. J Autoimmun 102:89–95PubMedCrossRef
170.
go back to reference Huynh M, Eggenhuizen PJ, Olson GL, Rao NB, Self CR, Sun Y et al (2019) HLA-DR15-specific inhibition attenuates autoreactivity to the Goodpasture antigen. J Autoimmun 103:102276PubMedCrossRef Huynh M, Eggenhuizen PJ, Olson GL, Rao NB, Self CR, Sun Y et al (2019) HLA-DR15-specific inhibition attenuates autoreactivity to the Goodpasture antigen. J Autoimmun 103:102276PubMedCrossRef
171.
go back to reference Stremska ME, Dai C, Venkatadri R, Wang H, Sabapathy V, Kumar G et al (2019) IL233, an IL-2-IL-33 hybrid cytokine induces prolonged remission of mouse lupus nephritis by targeting Treg cells as a single therapeutic agent. J Autoimmun 102:133–141PubMedPubMedCentralCrossRef Stremska ME, Dai C, Venkatadri R, Wang H, Sabapathy V, Kumar G et al (2019) IL233, an IL-2-IL-33 hybrid cytokine induces prolonged remission of mouse lupus nephritis by targeting Treg cells as a single therapeutic agent. J Autoimmun 102:133–141PubMedPubMedCentralCrossRef
172.
go back to reference Kolb H, Burkart V (2019) Chaperones may cause the focus of diabetes autoimmunity on distinct (pro)insulin peptides. J Autoimmun 105:102304PubMedCrossRef Kolb H, Burkart V (2019) Chaperones may cause the focus of diabetes autoimmunity on distinct (pro)insulin peptides. J Autoimmun 105:102304PubMedCrossRef
173.
go back to reference Tenspolde M, Zimmermann K, Weber LC, Hapke M, Lieber M, Dywicki J et al (2019) Regulatory T cells engineered with a novel insulin-specific chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes. J Autoimmun 103:102289PubMedCrossRef Tenspolde M, Zimmermann K, Weber LC, Hapke M, Lieber M, Dywicki J et al (2019) Regulatory T cells engineered with a novel insulin-specific chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes. J Autoimmun 103:102289PubMedCrossRef
174.
go back to reference Simakou T, Butcher JP, Reid S, Henriquez FL (2019) Alopecia areata: a multifactorial autoimmune condition. J Autoimmun 98:74–85PubMedCrossRef Simakou T, Butcher JP, Reid S, Henriquez FL (2019) Alopecia areata: a multifactorial autoimmune condition. J Autoimmun 98:74–85PubMedCrossRef
175.
go back to reference Erdei E, Shuey C, Pacheco B, Cajero M, Lewis J, Rubin RL (2019) Elevated autoimmunity in residents living near abandoned uranium mine sites on the Navajo Nation. J Autoimmun 99:15–23PubMedPubMedCentralCrossRef Erdei E, Shuey C, Pacheco B, Cajero M, Lewis J, Rubin RL (2019) Elevated autoimmunity in residents living near abandoned uranium mine sites on the Navajo Nation. J Autoimmun 99:15–23PubMedPubMedCentralCrossRef
176.
go back to reference Meier HCS, Parks CG, Liu HB, Sandler DP, Simonsick EM, Deane K et al (2019) Cellular aging over 13 years associated with incident antinuclear antibody positivity in the Baltimore Longitudinal Study of Aging. J Autoimmun 105:102295PubMedCrossRefPubMedCentral Meier HCS, Parks CG, Liu HB, Sandler DP, Simonsick EM, Deane K et al (2019) Cellular aging over 13 years associated with incident antinuclear antibody positivity in the Baltimore Longitudinal Study of Aging. J Autoimmun 105:102295PubMedCrossRefPubMedCentral
177.
go back to reference Essouma M, Nkeck JR, Endomba FT, Bigna JJ, Singwe-Ngandeu M, Hachulla E (2020) Systemic lupus erythematosus in Native sub-Saharan Africans: a systematic review and meta-analysis. J Autoimmun 106:102348PubMedCrossRef Essouma M, Nkeck JR, Endomba FT, Bigna JJ, Singwe-Ngandeu M, Hachulla E (2020) Systemic lupus erythematosus in Native sub-Saharan Africans: a systematic review and meta-analysis. J Autoimmun 106:102348PubMedCrossRef
178.
go back to reference Tanner S, Dufault B, Smolik I, Meng X, Anaparti V, Hitchon C et al (2019) A prospective study of the development of inflammatory arthritis in the family members of indigenous North American people with rheumatoid arthritis. Arthritis Rheum 71:1494–1503CrossRef Tanner S, Dufault B, Smolik I, Meng X, Anaparti V, Hitchon C et al (2019) A prospective study of the development of inflammatory arthritis in the family members of indigenous North American people with rheumatoid arthritis. Arthritis Rheum 71:1494–1503CrossRef
179.
go back to reference Desai RJ, Jin Y, Franklin PD, Lee YC, Bateman BT, Lii J et al (2019) Association of geography and access to health care providers with long-term prescription opioid use in Medicare patients with severe osteoarthritis: a cohort study. Arthritis Rheum 71:712–721CrossRef Desai RJ, Jin Y, Franklin PD, Lee YC, Bateman BT, Lii J et al (2019) Association of geography and access to health care providers with long-term prescription opioid use in Medicare patients with severe osteoarthritis: a cohort study. Arthritis Rheum 71:712–721CrossRef
180.
go back to reference Kronzer VL, Crowson CS, Sparks JA, Vassallo R, Davis JM 3rd. (2019) Investigating asthma, allergic disease, passive smoke exposure, and risk of rheumatoid arthritis. Arthritis Rheum 71:1217–1224CrossRef Kronzer VL, Crowson CS, Sparks JA, Vassallo R, Davis JM 3rd. (2019) Investigating asthma, allergic disease, passive smoke exposure, and risk of rheumatoid arthritis. Arthritis Rheum 71:1217–1224CrossRef
181.
go back to reference Liu X, Tedeschi SK, Lu B, Zaccardelli A, Speyer CB, Costenbader KH et al (2019) Long-term pysical activity and subsequent risk for rheumatoid arthritis among women: a prospective cohort study. Arthritis Rheum 71:1460–1471CrossRef Liu X, Tedeschi SK, Lu B, Zaccardelli A, Speyer CB, Costenbader KH et al (2019) Long-term pysical activity and subsequent risk for rheumatoid arthritis among women: a prospective cohort study. Arthritis Rheum 71:1460–1471CrossRef
182.
go back to reference Lee YC, Kremer J, Guan H, Greenberg J, Solomon DH (2019) Chronic opioid use in rheumatoid arthritis: prevalence and predictors. Arthritis Rheum 71:670–677CrossRef Lee YC, Kremer J, Guan H, Greenberg J, Solomon DH (2019) Chronic opioid use in rheumatoid arthritis: prevalence and predictors. Arthritis Rheum 71:670–677CrossRef
183.
go back to reference Chang C, Gershwin ME (2019) The myth of mycotoxins and mold injury. Clin Rev Allergy Immunol 57:449–455PubMedCrossRef Chang C, Gershwin ME (2019) The myth of mycotoxins and mold injury. Clin Rev Allergy Immunol 57:449–455PubMedCrossRef
184.
go back to reference Yamamoto E, Jorgensen TN (2019) Immunological effects of vitamin D and their relations to autoimmunity. J Autoimmun 100:7–16PubMedCrossRef Yamamoto E, Jorgensen TN (2019) Immunological effects of vitamin D and their relations to autoimmunity. J Autoimmun 100:7–16PubMedCrossRef
185.
go back to reference Huang C, Yi X, Long H, Zhang G, Wu H, Zhao M et al (2020) Disordered cutaneous microbiota in systemic lupus erythematosus. J Autoimmun 108:102391PubMedCrossRef Huang C, Yi X, Long H, Zhang G, Wu H, Zhao M et al (2020) Disordered cutaneous microbiota in systemic lupus erythematosus. J Autoimmun 108:102391PubMedCrossRef
186.
go back to reference van der Meulen TA, Harmsen HJM, Vila AV, Kurilshikov A, Liefers SC, Zhernakova A et al (2019) Shared gut, but distinct oral microbiota composition in primary Sjogren's syndrome and systemic lupus erythematosus. J Autoimmun 97:77–87PubMedCrossRef van der Meulen TA, Harmsen HJM, Vila AV, Kurilshikov A, Liefers SC, Zhernakova A et al (2019) Shared gut, but distinct oral microbiota composition in primary Sjogren's syndrome and systemic lupus erythematosus. J Autoimmun 97:77–87PubMedCrossRef
187.
go back to reference Amaya-Uribe L, Rojas M, Azizi G, Anaya JM, Gershwin ME (2019) Primary immunodeficiency and autoimmunity: a comprehensive review. J Autoimmun 99:52–72PubMedCrossRef Amaya-Uribe L, Rojas M, Azizi G, Anaya JM, Gershwin ME (2019) Primary immunodeficiency and autoimmunity: a comprehensive review. J Autoimmun 99:52–72PubMedCrossRef
188.
go back to reference Jamee M, Moniri S, Zaki-Dizaji M, Olbrich P, Yazdani R, Jadidi-Niaragh F et al (2019) Clinical, immunological, and genetic features in patients with activated PI3K delta syndrome (APDS): a systematic review. Clin Rev Allergy Immunol Jamee M, Moniri S, Zaki-Dizaji M, Olbrich P, Yazdani R, Jadidi-Niaragh F et al (2019) Clinical, immunological, and genetic features in patients with activated PI3K delta syndrome (APDS): a systematic review. Clin Rev Allergy Immunol
189.
go back to reference Gokhale AS, Gangaplara A, Lopez-Occasio M, Thornton AM, Shevach EM (2019) Selective deletion of Eos (Ikzf4) in T-regulatory cells leads to loss of suppressive function and development of systemic autoimmunity. J Autoimmun 105:102300PubMedCrossRef Gokhale AS, Gangaplara A, Lopez-Occasio M, Thornton AM, Shevach EM (2019) Selective deletion of Eos (Ikzf4) in T-regulatory cells leads to loss of suppressive function and development of systemic autoimmunity. J Autoimmun 105:102300PubMedCrossRef
190.
go back to reference Lourou N, Gavriilidis M, Kontoyiannis DL (2019) Lessons from studying the AU-rich elements in chronic inflammation and autoimmunity. J Autoimmun 104:102334PubMedCrossRef Lourou N, Gavriilidis M, Kontoyiannis DL (2019) Lessons from studying the AU-rich elements in chronic inflammation and autoimmunity. J Autoimmun 104:102334PubMedCrossRef
191.
go back to reference Lundtoft C, Seyfarth J, Oberstrass S, Rosenbauer J, Baechle C, Roden M et al (2019) Autoimmunity risk- and protection-associated IL7RA genetic variants differentially affect soluble and membrane IL-7R alpha expression. J Autoimmun 97:40–47PubMedCrossRef Lundtoft C, Seyfarth J, Oberstrass S, Rosenbauer J, Baechle C, Roden M et al (2019) Autoimmunity risk- and protection-associated IL7RA genetic variants differentially affect soluble and membrane IL-7R alpha expression. J Autoimmun 97:40–47PubMedCrossRef
192.
go back to reference Morianos I, Papadopoulou G, Semitekolou M, Xanthou G (2019) Activin-A in the regulation of immunity in health and disease. J Autoimmun 104:102314PubMedCrossRef Morianos I, Papadopoulou G, Semitekolou M, Xanthou G (2019) Activin-A in the regulation of immunity in health and disease. J Autoimmun 104:102314PubMedCrossRef
193.
go back to reference Pacheco Y, Acosta-Ampudia Y, Monsalve DM, Chang C, Gershwin ME, Anaya JM (2019) Bystander activation and autoimmunity. J Autoimmun 103:102301PubMedCrossRef Pacheco Y, Acosta-Ampudia Y, Monsalve DM, Chang C, Gershwin ME, Anaya JM (2019) Bystander activation and autoimmunity. J Autoimmun 103:102301PubMedCrossRef
194.
go back to reference Saferding V, Bluml S (2020) Innate immunity as the trigger of systemic autoimmune diseases. J Autoimmun 110:102382PubMedCrossRef Saferding V, Bluml S (2020) Innate immunity as the trigger of systemic autoimmune diseases. J Autoimmun 110:102382PubMedCrossRef
195.
go back to reference Vlachiotis S, Andreakos E (2019) Lambda interferons in immunity and autoimmunity. J Autoimmun 104:102319PubMedCrossRef Vlachiotis S, Andreakos E (2019) Lambda interferons in immunity and autoimmunity. J Autoimmun 104:102319PubMedCrossRef
196.
go back to reference Yao L, Zhou L, Xuan Y, Zhang P, Wang X, Wang T et al (2019) The proteasome activator REG gamma counteracts immunoproteasome expression and autoimmunity. J Autoimmun 103:102282PubMedCrossRef Yao L, Zhou L, Xuan Y, Zhang P, Wang X, Wang T et al (2019) The proteasome activator REG gamma counteracts immunoproteasome expression and autoimmunity. J Autoimmun 103:102282PubMedCrossRef
197.
go back to reference Chen B, Vousden KA, Naiman B, Turman S, Sun H, Wang S et al (2019) Humanised effector-null Fc gamma RIIA antibody inhibits immune complex-mediated proinflammatory responses. Ann Rheum Dis 78:228–237PubMedCrossRef Chen B, Vousden KA, Naiman B, Turman S, Sun H, Wang S et al (2019) Humanised effector-null Fc gamma RIIA antibody inhibits immune complex-mediated proinflammatory responses. Ann Rheum Dis 78:228–237PubMedCrossRef
198.
go back to reference Stohl W, Yu N, Chalmers S, Putterman C, Jacob CO (2020) Development of murine systemic lupus erythematosus in the absence of BAFF. Arthritis Rheum 72:292–302CrossRef Stohl W, Yu N, Chalmers S, Putterman C, Jacob CO (2020) Development of murine systemic lupus erythematosus in the absence of BAFF. Arthritis Rheum 72:292–302CrossRef
199.
go back to reference Xiao N, Wei J, Xu S, Du H, Huang M, Zhang S et al (2019) cGAS activation causes lupus-like autoimmune disorders in a TREX1 mutant mouse model. J Autoimmun 100:84–94PubMedCrossRef Xiao N, Wei J, Xu S, Du H, Huang M, Zhang S et al (2019) cGAS activation causes lupus-like autoimmune disorders in a TREX1 mutant mouse model. J Autoimmun 100:84–94PubMedCrossRef
200.
go back to reference Wade SM, Trenkmann M, McGarry T, Canavan M, Marzaioli V, Wade SC et al (2019) Altered expression of microRNA-23a in psoriatic arthritis modulates synovial fibroblast pro-inflammatory mechanisms via phosphodiesterase 4B. J Autoimmun 96:86–93PubMedCrossRef Wade SM, Trenkmann M, McGarry T, Canavan M, Marzaioli V, Wade SC et al (2019) Altered expression of microRNA-23a in psoriatic arthritis modulates synovial fibroblast pro-inflammatory mechanisms via phosphodiesterase 4B. J Autoimmun 96:86–93PubMedCrossRef
201.
go back to reference Mustelin T, Bottini N, Stanford SM (2019) The contribution of PTPN22 to rheumatic disease. Arthritis Rheum 71:486–495CrossRef Mustelin T, Bottini N, Stanford SM (2019) The contribution of PTPN22 to rheumatic disease. Arthritis Rheum 71:486–495CrossRef
202.
go back to reference Tison A, Quere G, Misery L, Funck-Brentano E, Danlos FX, Routier E et al (2019) Safety and efficacy of immune checkpoint inhibitors in patients with cancer and preexisting autoimmune disease: a nationwide, multicenter cohort study. Arthritis Rheum 71:2100–2111CrossRef Tison A, Quere G, Misery L, Funck-Brentano E, Danlos FX, Routier E et al (2019) Safety and efficacy of immune checkpoint inhibitors in patients with cancer and preexisting autoimmune disease: a nationwide, multicenter cohort study. Arthritis Rheum 71:2100–2111CrossRef
203.
go back to reference Alissafi T, Hatzioannou A, Legaki AI, Varveri A, Verginis P (2019) Balancing cancer immunotherapy and immune-related adverse events: the emerging role of regulatory T cells. J Autoimmun 104:102310PubMedCrossRef Alissafi T, Hatzioannou A, Legaki AI, Varveri A, Verginis P (2019) Balancing cancer immunotherapy and immune-related adverse events: the emerging role of regulatory T cells. J Autoimmun 104:102310PubMedCrossRef
204.
go back to reference Thiel J, Alter C, Luppus S, Eckstein A, Tan S, Fuhrer D et al (2019) MicroRNA-183 and microRNA-96 are associated with autoimmune responses by regulating T cell activation. J Autoimmun 96:94–103PubMedCrossRef Thiel J, Alter C, Luppus S, Eckstein A, Tan S, Fuhrer D et al (2019) MicroRNA-183 and microRNA-96 are associated with autoimmune responses by regulating T cell activation. J Autoimmun 96:94–103PubMedCrossRef
205.
go back to reference Jamilloux Y, El Jammal T, Vuitton L, Gerfaud-Valentin M, Kerever S, Seve P (2019) JAK inhibitors for the treatment of autoimmune and inflammatory diseases. Autoimmun Rev 18:102390PubMedCrossRef Jamilloux Y, El Jammal T, Vuitton L, Gerfaud-Valentin M, Kerever S, Seve P (2019) JAK inhibitors for the treatment of autoimmune and inflammatory diseases. Autoimmun Rev 18:102390PubMedCrossRef
Metadata
Title
Autoimmunity in 2019
Author
Carlo Selmi
Publication date
01-12-2020
Publisher
Springer US
Published in
Clinical Reviews in Allergy & Immunology / Issue 3/2020
Print ISSN: 1080-0549
Electronic ISSN: 1559-0267
DOI
https://doi.org/10.1007/s12016-020-08808-3

Other articles of this Issue 3/2020

Clinical Reviews in Allergy & Immunology 3/2020 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine