The role of stroma and epithelial cells in primary Sjögren’s syndrome

Saba Asam1, Georgiana Neag1, Onorina Berardicurti1, David H. Gardner1, Francesca Barone1
1Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK

Tóm tắt

Từ khóa


Tài liệu tham khảo

Manoussakis, 2007, The role of epithelial cells in the pathogenesis of Sjögren’s syndrome, Clin Rev Allergy Immunol, 32, 225, 10.1007/s12016-007-8007-4

Fox, 2012, The salivary gland epithelial cell in Sjögren’s syndrome: what are the steps involved in wounding or killing their secretory function?, J Rheumatol, 39, 1117, 10.3899/jrheum.120278

Mitsias, 2006, Sjögren’s syndrome: why autoimmune epithelitis?, Oral Dis, 12, 523, 10.1111/j.1601-0825.2006.01292.x

Qin, 2015, Epidemiology of primary Sjögren’s syndrome: a systematic review and meta-analysis, Ann Rheum Dis, 74, 1983, 10.1136/annrheumdis-2014-205375

Maldini, 2014, Epidemiology of primary Sjögren’s syndrome in a French multiracial/multiethnic area, Arthritis Care Res, 66, 454, 10.1002/acr.22115

Reveille, 1984, Primary Sjögren’s syndrome and other autoimmune diseases in families. Prevalence and immunogenetic studies in six kindreds, Ann Intern Med, 101, 748, 10.7326/0003-4819-101-6-748

Cruz-Tapias, 2012, HLA and Sjögren’s syndrome susceptibility. A meta-analysis of worldwide studies, Autoimmun Rev, 11, 281, 10.1016/j.autrev.2011.10.002

Lessard, 2013, Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjögren’s syndrome, Nat Genet, 45, 1284, 10.1038/ng.2792

Takaoka, 2005, Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors, Nature, 434, 243, 10.1038/nature03308

Theander, 2011, Lymphoid organisation in labial salivary gland biopsies is a possible predictor for the development of malignant lymphoma in primary Sjögren’s syndrome, Ann Rheum Dis, 70, 1363, 10.1136/ard.2010.144782

Yao, 2013, Type I interferons in Sjögren’s syndrome, Autoimmun Rev, 12, 558, 10.1016/j.autrev.2012.10.006

Imgenberg-Kreuz, 2018, Epigenetic alterations in primary Sjögren’s syndrome – an overview, Clin Immunol, 196, 12, 10.1016/j.clim.2018.04.004

Alevizos, 2011, MicroRNA expression profiles as biomarkers of minor salivary gland inflammation and dysfunction in Sjögren’s syndrome, Arthritis Rheum, 63, 535, 10.1002/art.30131

Chen, 2016, The role of microRNAs in the pathogenesis of autoimmune diseases, Autoimmun Rev, 15, 1171, 10.1016/j.autrev.2016.09.003

Thabet, 2013, Epigenetic dysregulation in salivary glands from patients with primary Sjögren’s syndrome may be ascribed to infiltrating B cells, J Autoimmun, 41, 175, 10.1016/j.jaut.2013.02.002

Lucchesi, 2014, EBV and other viruses as triggers of tertiary lymphoid structures in primary Sjögren’s syndrome, Expert Rev Clin Immunol, 10, 445, 10.1586/1744666X.2014.892417

Tzioufas, 1999, Autoantibodies to La/SSB in patients with primary Sjögren’s syndrome (pSS) are associated with upregulation of La/SSB mRNA in minor salivary gland biopsies (MSGs), J Autoimmun, 13, 429, 10.1006/jaut.1999.0333

Gong, 2014, Differentiation of follicular helper T cells by salivary gland epithelial cells in primary Sjögren’s syndrome, J Autoimmun, 51, 57, 10.1016/j.jaut.2013.11.003

Goules, 2017, Insight into pathogenesis of Sjögren’s syndrome: dissection on autoimmune infiltrates and epithelial cells, Clin Immunol, 182, 30, 10.1016/j.clim.2017.03.007

Kong, 1998, Inappropriate apoptosis of salivary and lacrimal gland epithelium of immunodeficient NOD-scid mice, Clin Exp Rheumatol, 16, 675

Manoussakis, 2010, The role of intrinsic epithelial activation in the pathogenesis of Sjögren’s syndrome, J Autoimmun, 35, 219, 10.1016/j.jaut.2010.06.011

Szyszko, 2011, Salivary glands of primary Sjögren’s syndrome patients express factors vital for plasma cell survival, Arthritis Res Ther, 13, 10.1186/ar3220

Szyszko, 2011, Phenotypic diversity of peripheral blood plasma cells in primary Sjögren’s syndrome, Scand J Immunol, 73, 18, 10.1111/j.1365-3083.2010.02475.x

Lisi, 2015, Co-culture system of human salivary gland epithelial cells and immune cells from primary Sjögren’s syndrome patients: an in vitro approach to study the effects of Rituximab on the activation of the Raf-1/ERK1/2 pathway, Int Immunol, 27, 183, 10.1093/intimm/dxu100

Shi, 2018, PD-1 controls follicular T helper cell positioning and function, Immunity, 49, 264, 10.1016/j.immuni.2018.06.012

Barone, 2015, IL-22 regulates lymphoid chemokine production and assembly of tertiary lymphoid organs, Proc Natl Acad Sci U S A, 112, 11024, 10.1073/pnas.1503315112

Gordon, 2001, Autoantibodies in primary Sjögren’s syndrome: new insights into mechanisms of autoantibody diversification and disease pathogenesis, Autoimmunity, 34, 123, 10.3109/08916930109001960

Konsta, 2014, The contribution of epigenetics in Sjögren’s Syndrome, Front Genet, 5, 71, 10.3389/fgene.2014.00071

Imgenberg-Kreuz, 2016, Genome-wide DNA methylation analysis in multiple tissues in primary Sjögren’s syndrome reveals regulatory effects at interferon-induced genes, Ann Rheum Dis, 75, 2029, 10.1136/annrheumdis-2015-208659

Bordron, 2018, Influence of epigenetic in Sjögren’s syndrome, Rev Med Interne, 39, 346, 10.1016/j.revmed.2017.09.003

Li, 2013, Interferons in Sjögren’s syndrome: genes, mechanisms, and effects, Front Immunol, 4, 290, 10.3389/fimmu.2013.00290

Carbone, 2000, Pathological features of lymphoid proliferations of the salivary glands: lymphoepithelial sialadenitis versus low-grade B-cell lymphoma of the malt type, Ann Otol Rhinol Laryngol, 109, 1170, 10.1177/000348940010901217

Ruddle, 2016, High endothelial venules and lymphatic vessels in tertiary lymphoid organs: characteristics, functions, and regulation, Front Immunol, 7, 491, 10.3389/fimmu.2016.00491

Salomonsson, 2003, Cellular basis of ectopic germinal center formation and autoantibody production in the target organ of patients with Sjögren’s syndrome, Arthritis Rheum, 48, 3187, 10.1002/art.11311

Aziz, 1992, Vascular endothelium and lymphocyte adhesion molecules in minor salivary glands of patients with Sjögren’s syndrome, J Clin Lab Immunol, 37, 39

Marchetti, 1989, Microscopic and ultrastructural study of the lymphatic system in the human parotid gland, Acta Anat, 134, 106, 10.1159/000146673

Ruddle, 2014, Lymphatic vessels and tertiary lymphoid organs, J Clin Invest, 124, 953, 10.1172/JCI71611

Petrova, 2018, Organ-specific lymphatic vasculature: from development to pathophysiology, J Exp Med, 215, 35, 10.1084/jem.20171868

Saito, 1993, Expression of cell adhesion molecules in the salivary and lacrimal glands of Sjögren’s syndrome, J Clin Lab Anal, 7, 180, 10.1002/jcla.1860070309

Reichardt, 2013, A role for LFA-1 in delaying T-lymphocyte egress from lymph nodes, Embo J, 32, 829, 10.1038/emboj.2013.33

Mikulowska-Mennis, 2001, Lymphocyte migration to inflamed lacrimal glands is mediated by vascular cell adhesion molecule-1/α4β1 integrin, peripheral node addressin/l-selectin, and lymphocyte function–associated antigen-1 adhesion pathways, Am J Pathol, 159, 671, 10.1016/S0002-9440(10)61738-5

Park, 2010, Distinct roles for LFA-1 affinity regulation during T-cell adhesion, diapedesis, and interstitial migration in lymph nodes, Blood, 115, 1572, 10.1182/blood-2009-08-237917

Hansen, 2005, Dysregulation of chemokine receptor expression and function by B cells of patients with primary Sjögren’s syndrome, Arthritis Rheum, 52, 2109, 10.1002/art.21129

Ley, 2007, Getting to the site of inflammation: the leukocyte adhesion cascade updated, Nat Rev Immunol, 7, 678, 10.1038/nri2156

Stranford, 2012, Follicular dendritic cells, conduits, lymphatic vessels, and high endothelial venules in tertiary lymphoid organs: parallels with lymph node stroma, Front Immunol, 3, 350, 10.3389/fimmu.2012.00350

Bartoloni, 2015, Characterization of circulating endothelial microparticles and endothelial progenitor cells in primary Sjögren’s syndrome: new markers of chronic endothelial damage?, Rheumatology, 54, 536, 10.1093/rheumatology/keu320

Aziz, 1996, Expression of selectins (CD62 E, L, P) and cellular adhesion molecules in primary Sjögren’s syndrome: questions to immunoregulation, Clin Immunol Immunopathol, 80, 55, 10.1006/clin.1996.0094

Turkcapar, 2005, Vasculitis and expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin in salivary glands of patients with Sjögren’s syndrome, J Rheumatol, 32, 1063

Daniels, 1984, Labial salivary gland biopsy in Sjögren’s syndrome. Assessment as a diagnostic criterion in 362 suspected cases, Arthritis Rheum, 27, 147, 10.1002/art.1780270205

Amft, 2001, Ectopic expression of the B cell–attracting chemokine BCA-1 (CXCL13) on endothelial cells and within lymphoid follicles contributes to the establishment of germinal center–like structures in Sjögren’s syndrome, Arthritis Rheum, 44, 2633, 10.1002/1529-0131(200111)44:11<2633::AID-ART443>3.0.CO;2-9

Bombardieri, 2012, Ectopic lymphoid neogenesis and lymphoid chemokines in Sjögren’s syndrome: at the interplay between chronic inflammation, autoimmunity and lymphomagenesis, Curr Pharm Biotechnol, 13, 1989, 10.2174/138920112802273209

McGettrick, 2009, Direct observations of the kinetics of migrating T cells suggest active retention by endothelial cells with continual bidirectional migration, J Leukoc Biol, 85, 98, 10.1189/jlb.0508301

Sisto, 2012, Neuropilin-1 is upregulated in Sjögren’s syndrome and contributes to pathological neovascularization, Histochem Cell Biol, 137, 669, 10.1007/s00418-012-0910-y

Daniels, 2011, Associations between salivary gland histopathologic diagnoses and phenotypic features of Sjögren’s syndrome among 1 726 registry participants, Arthritis Rheum, 63, 2021, 10.1002/art.30381

Nagy, 2008, Vascular permeability, vascular hyperpermeability and angiogenesis, Angiogenesis, 11, 109, 10.1007/s10456-008-9099-z

Nayar, 2016, Bimodal expansion of the lymphatic vessels is regulated by the sequential expression of IL-7 and lymphotoxin alpha1beta2 in newly formed tertiary lymphoid structures, J Immunol, 197, 1957, 10.4049/jimmunol.1500686

McCall, 2015, Characterization of angiogenesis and lymphangiogenesis in human minor salivary glands with Sjögren’s syndrome, J Histochem Cytochem, 63, 340, 10.1369/0022155415573323

Jonsson, 2007, Serological implications of germinal center–like structures in primary Sjögren’s syndrome, J Rheumatol, 34, 2044

Rosen, 1992, Brazen anatomy, Nature, 358, 463, 10.1038/358463b0

Cyster, 2000, Follicular stromal cells and lymphocyte homing to follicles, Immunol Rev, 176, 181, 10.1034/j.1600-065X.2000.00618.x

Onder, 2012, IL-7–producing stromal cells are critical for lymph node remodeling, Blood, 120, 4675, 10.1182/blood-2012-03-416859

Patel, 2014, Stroma: fertile soil for inflammation, Best Pract Res Clin Rheumatol, 28, 565, 10.1016/j.berh.2014.10.022

Parsonage, 2005, A stromal address code defined by fibroblasts, Trends Immunol, 26, 150, 10.1016/j.it.2004.11.014

Barone, 2016, Stromal fibroblasts in tertiary lymphoid structures: a novel target in chronic inflammation, Front Immunol, 7, 477, 10.3389/fimmu.2016.00477

Ferreira, 2013, Interactions between developing nerves and salivary glands, Organogenesis, 9, 199, 10.4161/org.25224

Konttinen, 1992, Peptide-containing nerves in labial salivary glands in Sjögren’s syndrome, Arthritis Rheum, 35, 815, 10.1002/art.1780350717

Pedersen, 2000, Innervation pattern and Ca2+ signalling in labial salivary glands of healthy individuals and patients with primary Sjögren’s syndrome (pSS), J Oral Pathol Med, 29, 97, 10.1034/j.1600-0714.2000.290301.x

Imrich, 2015, Predominant glandular cholinergic dysautonomia in patients with primary Sjögren’s syndrome, Arthritis Rheumatol, 67, 1345, 10.1002/art.39044

Batbayar, 2002, Changes of the nerve fibers innervating the minor salivary glands in Sjögren syndrome, Orv Hetil, 143, 1585

Passafaro, 2010, Cholinergic autoantibodies from primary Sjögren’s syndrome modulate submandibular gland Na+/K+-ATPase activity via prostaglandin E2 and cyclic AMP, Eur J Oral Sci, 118, 131, 10.1111/j.1600-0722.2010.00716.x

Enger, 2014, Calcium signaling and cell volume regulation are altered in Sjögren’s syndrome, Acta Odontol Scand, 72, 549, 10.3109/00016357.2013.879995

Jin, 2012, Autoantibodies in Sjögren’s syndrome patients acutely inhibit muscarinic receptor function, Oral Dis, 18, 132, 10.1111/j.1601-0825.2011.01853.x

Waterman, 2000, Inhibitory effects of muscarinic receptor autoantibodies on parasympathetic neurotransmission in Sjögren’s syndrome, Arthritis Rheum, 43, 1647, 10.1002/1529-0131(200007)43:7<1647::AID-ANR31>3.0.CO;2-P

Naito, 2005, Muscarinic acetylcholine receptor autoantibodies in patients with Sjögren’s syndrome, Ann Rheum Dis, 64, 510, 10.1136/ard.2004.025478

Baum, 2011, Fibroblasts and myofibroblasts: what are we talking about?, J Cardiovasc Pharmacol, 57, 376, 10.1097/FJC.0b013e3182116e39

Nolte, 2008, Diversity of fibroblasts—a review on implications for skin tissue engineering, Cells Tissues Organs, 187, 165, 10.1159/000111805

Tao, 2017, Cancer associated fibroblasts: an essential role in the tumor microenvironment, Oncol Lett, 14, 2611, 10.3892/ol.2017.6497

Kalluri, 2006, Fibroblasts in cancer, Nat Rev Cancer, 6, 392, 10.1038/nrc1877

Buckley, 2001, Fibroblasts regulate the switch from acute resolving to chronic persistent inflammation, Trends Immunol, 22, 199, 10.1016/S1471-4906(01)01863-4

Cyster, 1999, Chemokines and cell migration in secondary lymphoid organs, Science, 286, 2098, 10.1126/science.286.5447.2098

Grogan, 2012, A role for Th17 cells in the regulation of tertiary lymphoid follicles, Eur J Immunol, 42, 2255, 10.1002/eji.201242656

Peduto, 2009, Inflammation recapitulates the ontogeny of lymphoid stromal cells, J Immunol, 182, 5789, 10.4049/jimmunol.0803974

Zhang, 2016, Co-stimulatory and co-inhibitory pathways in autoimmunity, Immunity, 44, 1034, 10.1016/j.immuni.2016.04.017

2015, Introduction to costimulation and costimulatory molecules, Developing costimulatory molecules for immunotherapy of diseases, 1, 10.1016/B978-0-12-802585-7.00001-7

Randall, 2008, Development of secondary lymphoid organs, Annu Rev Immunol, 26, 627, 10.1146/annurev.immunol.26.021607.090257

Yang, 2000, CD21-mediated entry and stable infection by Epstein–Barr virus in canine and rat cells, J Virol, 74, 10745, 10.1128/JVI.74.22.10745-10751.2000

Yin, 2017, Editorial: tertiary lymphoid organs (TLOs): powerhouses of disease immunity, Front Immunol, 8, 228, 10.3389/fimmu.2017.00228

Nayar S . Lymphoid like stromal cells in a model of tertiary lymphoid organ formation. PhD Thesis, University of Birmingham, Birmingham, UK, 2014.

Dieu-Nosjean, 2008, Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures, J Clin Oncol, 26, 4410, 10.1200/JCO.2007.15.0284

Ciccia, 2017, Ectopic expression of CXCL13, BAFF, APRIL and LT-beta is associated with artery tertiary lymphoid organs in giant cell arteritis, Ann Rheum Dis, 76, 235, 10.1136/annrheumdis-2016-209217

Pikor, 2015, Meningeal tertiary lymphoid tissues and multiple sclerosis: a gathering place for diverse types of immune cells during CNS autoimmunity, Front Immunol, 6, 657

Kang, 2017, BAFF induces tertiary lymphoid structures and positions T cells within the glomeruli during lupus nephritis, J Immunol, 198, 2602, 10.4049/jimmunol.1600281

Weyand, 2003, Ectopic germinal center formation in rheumatoid synovitis, Ann N Y Acad Sci, 987, 140, 10.1111/j.1749-6632.2003.tb06042.x

Nocturne, 2018, B cells in the pathogenesis of primary Sjögren syndrome, Nat Rev Rheumatol, 14, 133, 10.1038/nrrheum.2018.1

Nocturne, 2015, Sjögren Syndrome–associated lymphomas: an update on pathogenesis and management, Br J Haematol, 168, 317, 10.1111/bjh.13192

Buckley, 2015, Stromal cells in chronic inflammation and tertiary lymphoid organ formation, Annu Rev Immunol, 33, 715, 10.1146/annurev-immunol-032713-120252

van de Pavert, 2010, New insights into the development of lymphoid tissues, Nat Rev Immunol, 10, 664, 10.1038/nri2832

Kumar, 2014, Biological function of activation-induced cytidine deaminase (AID), Biomed J, 37, 269, 10.4103/2319-4170.128734

Allen, 2007, Germinal-center organization and cellular dynamics, Immunity, 27, 190, 10.1016/j.immuni.2007.07.009

Royer, 1997, Lymphomas in patients with Sjögren’s syndrome are marginal zone B-cell neoplasms, arise in diverse extranodal and nodal sites, and are not associated with viruses, Blood, 90, 766, 10.1182/blood.V90.2.766

Victora, 2012, Germinal centers, Annu Rev Immunol, 30, 429, 10.1146/annurev-immunol-020711-075032

Risselada, 2014, The prognostic value of routinely performed minor salivary gland assessments in primary Sjögren’s syndrome, Ann Rheum Dis, 73, 1537, 10.1136/annrheumdis-2013-204634

Risselada, 2013, The role of ectopic germinal centers in the immunopathology of primary Sjögren’s syndrome: a systematic review, Semin Arthritis Rheum, 42, 368, 10.1016/j.semarthrit.2012.07.003

Haacke, 2017, Germinal centres in diagnostic labial gland biopsies of patients with primary Sjögren’s syndrome are not predictive for parotid MALT lymphoma development, Ann Rheum Dis, 76, 1781, 10.1136/annrheumdis-2017-211290

Oni, 2016, Eligibility for clinical trials in primary Sjögren’s syndrome: lessons from the UK Primary Sjögren’s Syndrome Registry, Rheumatology, 55, 544

Bombardieri, 2012, Inducible tertiary lymphoid structures, autoimmunity, and exocrine dysfunction in a novel model of salivary gland inflammation in C57BL/6 mice, J Immunol, 189, 3767, 10.4049/jimmunol.1201216

Franks, 2016, Targeting B cells in treatment of autoimmunity, Curr Opin Immunol, 43, 39, 10.1016/j.coi.2016.09.003

Hamza, 2012, Persistence of immunoglobulin-producing cells in parotid salivary glands of patients with primary Sjögren’s syndrome after B cell depletion therapy, Ann Rheum Dis, 71, 1881, 10.1136/annrheumdis-2011-201189

Barone, 2005, Association of CXCL13 and CCL21 expression with the progressive organization of lymphoid-like structures in Sjögren’s syndrome, Arthritis Rheum, 52, 1773, 10.1002/art.21062

Hu, 2015, Artery tertiary lymphoid organs control aorta immunity and protect against atherosclerosis via vascular smooth muscle cell lymphotoxin beta receptors, Immunity, 42, 1100, 10.1016/j.immuni.2015.05.015