Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation
Abstract
:1. Introduction
2. Innate Immune Cells in pSS
2.1. Dendritic Cells (DCs)
2.2. Macrophages
2.3. Mast Cells
2.4. Salivary Gland Epithelial Cells (SGECs)
2.5. Endothelial Cells
2.6. Mucosa-Associated Invariant T (MAIT) Cells
2.7. Natural Killer Cells (NKs) and Natural Killer T Cells (NKTs)
2.8. Innate Lymphoid Cells (ILCs)
3. Main Mechanisms of Inflammation in pSS Related to Innate Immunity
3.1. IFN Signature, TLRs Activation, Intracellular Pathways Depending on JAK/STAT and MAP/ERK
3.2. Inflammasome
3.3. Non Coding RNAs (ncRNAs) and Their Epigenetic Effect
4. Conclusions
5. Future Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Mariette, X.; Criswell, L.A. Primary Sjögren’s Syndrome. N. Engl. J. Med. 2018, 378, 931–939. [Google Scholar] [CrossRef] [PubMed]
- Johnsen, S.J.; Brun, J.G.; Goransson, L.G.; Smastuen, M.C.; Johannesen, T.B.; Haldorsen, K.; Harboe, E.; Jonsson, R.; Meyer, P.A.; Omdal, R. Risk of non-Hodgkin’s lymphoma in primary Sjogren’s syndrome: A population-based study. Arthritis Care Res. 2013, 65, 816–821. [Google Scholar] [CrossRef] [PubMed]
- Patel, R.; Shahane, A. The epidemiology of Sjogren’s syndrome. Clin. Epidemiol. 2014, 6, 247–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Voulgarelis, M.; Tzioufas, A.G. Pathogenetic mechanisms in the initiation and perpetuation of Sjogren’s syndrome. Nat. Rev. Rheumatol. 2010, 6, 529–537. [Google Scholar] [CrossRef] [PubMed]
- Kiripolsky, J.; McCabe, L.G.; Kramer, J.M. Innate immunity in Sjogren’s syndrome. Clin. Immunol. 2017, 182, 4–13. [Google Scholar] [CrossRef] [PubMed]
- Kawai, T.; Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011, 34, 637–650. [Google Scholar] [CrossRef] [Green Version]
- Low, H.Z.; Witte, T. Aspects of innate immunity in Sjogren’s syndrome. Arthritis Res. 2011, 13, 218. [Google Scholar] [CrossRef] [Green Version]
- Bombardieri, M.; Pitzalis, C. Ectopic lymphoid neogenesis and lymphoid chemokines in Sjogren’s syndrome: At the interplay between chronic inflammation, autoimmunity and lymphomagenesis. Curr. Pharm. Biotechnol. 2012, 13, 1989–1996. [Google Scholar] [CrossRef]
- Hillen, M.R.; Pandit, A.; Blokland, S.L.M.; Hartgring, S.A.Y.; Bekker, C.P.J.; van der Heijden, E.H.M.; Servaas, N.H.; Rossato, M.; Kruize, A.A.; van Roon, J.A.G.; et al. Plasmacytoid DCs From Patients With Sjogren’s Syndrome Are Transcriptionally Primed for Enhanced Pro-inflammatory Cytokine Production. Front. Immunol. 2019, 10, 2096. [Google Scholar] [CrossRef] [Green Version]
- Ozaki, Y.; Ito, T.; Son, Y.; Amuro, H.; Shimamoto, K.; Sugimoto, H.; Katashiba, Y.; Ogata, M.; Miyamoto, R.; Murakami, N.; et al. Decrease of blood dendritic cells and increase of tissue-infiltrating dendritic cells are involved in the induction of Sjogren’s syndrome but not in the maintenance. Clin. Exp. Immunol. 2010, 159, 315–326. [Google Scholar] [CrossRef]
- Ainola, M.; Porola, P.; Takakubo, Y.; Przybyla, B.; Kouri, V.P.; Tolvanen, T.A.; Hanninen, A.; Nordstrom, D.C. Activation of plasmacytoid dendritic cells by apoptotic particles—Mechanism for the loss of immunological tolerance in Sjogren’s syndrome. Clin. Exp. Immunol. 2018, 191, 301–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swiecki, M.; Colonna, M. Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance. Immunol. Rev. 2010, 234, 142–162. [Google Scholar] [CrossRef] [PubMed]
- Swiecki, M.; Colonna, M. The multifaceted biology of plasmacytoid dendritic cells. Nat. Rev. Immunol. 2015, 15, 471–485. [Google Scholar] [CrossRef] [PubMed]
- Bave, U.; Nordmark, G.; Lovgren, T.; Ronnelid, J.; Cajander, S.; Eloranta, M.L.; Alm, G.V.; Ronnblom, L. Activation of the type I interferon system in primary Sjogren’s syndrome: A possible etiopathogenic mechanism. Arthritis Rheum. 2005, 52, 1185–1195. [Google Scholar] [CrossRef] [PubMed]
- Vakaloglou, K.M.; Mavragani, C.P. Activation of the type I interferon pathway in primary Sjogren’s syndrome: An update. Curr. Opin. Rheumatol. 2011, 23, 459–464. [Google Scholar] [CrossRef]
- El Shikh, M.E.; Pitzalis, C. Follicular dendritic cells in health and disease. Front. Immunol. 2012, 3, 292. [Google Scholar] [CrossRef] [Green Version]
- Aloisi, F.; Pujol-Borrell, R. Lymphoid neogenesis in chronic inflammatory diseases. Nat. Rev. Immunol. 2006, 6, 205–217. [Google Scholar] [CrossRef]
- Lambert, N.C. Nonendocrine mechanisms of sex bias in rheumatic diseases. Nat. Rev. Rheumatol. 2019, 15, 673–686. [Google Scholar] [CrossRef]
- Stout, R.D.; Jiang, C.; Matta, B.; Tietzel, I.; Watkins, S.K.; Suttles, J. Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences. J. Immunol. 2005, 175, 342–349. [Google Scholar] [CrossRef]
- Christodoulou, M.I.; Kapsogeorgou, E.K.; Moutsopoulos, H.M. Characteristics of the minor salivary gland infiltrates in Sjogren’s syndrome. J. Autoimmun. 2010, 34, 400–407. [Google Scholar] [CrossRef]
- Kinoshita, S.; Nakamura, T.; Nishida, K. Pathological keratinization of ocular surface epithelium. Adv. Exp. Med. Biol. 2002, 506, 641–646. [Google Scholar] [CrossRef] [PubMed]
- McNamara, N.A. Molecular mechanisms of keratinizing ocular surface disease. Optom. Vis. Sci. 2010, 87, 233–238. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Yu, Y.F.; Liu, C.H.; Wang, C.M. Significance of M2 macrophage in tubulointerstitial disease secondary to primary Sjogren’s disease. Ren. Fail. 2018, 40, 634–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skopouli, F.N.; Li, L.; Boumba, D.; Stefanaki, S.; Hanel, K.; Moutsopoulos, H.M.; Krilis, S.A. Association of mast cells with fibrosis and fatty infiltration in the minor salivary glands of patients with Sjogren’s syndrome. Clin. Exp. Rheumatol. 1998, 16, 63–65. [Google Scholar]
- Perez, P.; Goicovich, E.; Alliende, C.; Aguilera, S.; Leyton, C.; Molina, C.; Pinto, R.; Romo, R.; Martinez, B.; Gonzalez, M.J. Differential expression of matrix metalloproteinases in labial salivary glands of patients with primary Sjogren’s syndrome. Arthritis Rheum. 2000, 43, 2807–2817. [Google Scholar] [CrossRef]
- Dinescu, S.C.; ForTofoiu, M.C.; Bumbea, A.M.; Ciurea, P.L.; Busuioc, C.J.; Musetescu, A.E. Histopathological and immunohistochemical profile in primary Sjogren’s syndrome. Rom. J. Morphol. Embryol. 2017, 58, 409–417. [Google Scholar]
- Barrera, M.J.; Bahamondes, V.; Sepulveda, D.; Quest, A.F.; Castro, I.; Cortes, J.; Aguilera, S.; Urzua, U.; Molina, C.; Perez, P.; et al. Sjogren’s syndrome and the epithelial target: A comprehensive review. J. Autoimmun. 2013, 42, 7–18. [Google Scholar] [CrossRef]
- Van Itallie, C.M.; Anderson, J.M. Architecture of tight junctions and principles of molecular composition. Semin. Cell Dev. Biol. 2014, 36, 157–165. [Google Scholar] [CrossRef] [Green Version]
- Baker, O.J.; Camden, J.M.; Redman, R.S.; Jones, J.E.; Seye, C.I.; Erb, L.; Weisman, G.A. Proinflammatory cytokines tumor necrosis factor-alpha and interferon-gamma alter tight junction structure and function in the rat parotid gland Par-C10 cell line. Am. J. Physiol. Cell Physiol. 2008, 295, C1191–C1201. [Google Scholar] [CrossRef] [Green Version]
- Ewert, P.; Aguilera, S.; Alliende, C.; Kwon, Y.J.; Albornoz, A.; Molina, C.; Urzua, U.; Quest, A.F.; Olea, N.; Perez, P.; et al. Disruption of tight junction structure in salivary glands from Sjogren’s syndrome patients is linked to proinflammatory cytokine exposure. Arthritis Rheum. 2010, 62, 1280–1289. [Google Scholar] [CrossRef]
- Fox, R.I. The salivary gland epithelial cell in Sjogren’s Syndrome: What are the steps involved in wounding or killing their secretory function? J. Rheumatol. 2012, 39, 1117–1119. [Google Scholar] [CrossRef] [Green Version]
- Manoussakis, M.N.; Kapsogeorgou, E.K. The role of epithelial cells in the pathogenesis of Sjogren’s syndrome. Clin. Rev. Allergy Immunol. 2007, 32, 225–230. [Google Scholar] [CrossRef]
- Gong, Y.Z.; Nititham, J.; Taylor, K.; Miceli-Richard, C.; Sordet, C.; Wachsmann, D.; Bahram, S.; Georgel, P.; Criswell, L.A.; Sibilia, J.; et al. Differentiation of follicular helper T cells by salivary gland epithelial cells in primary Sjogren’s syndrome. J. Autoimmun. 2014, 51, 57–66. [Google Scholar] [CrossRef] [PubMed]
- Manoussakis, M.N.; Kapsogeorgou, E.K. The role of intrinsic epithelial activation in the pathogenesis of Sjogren’s syndrome. J. Autoimmun. 2010, 35, 219–224. [Google Scholar] [CrossRef] [PubMed]
- Mitsias, D.I.; Kapsogeorgou, E.K.; Moutsopoulos, H.M. The role of epithelial cells in the initiation and perpetuation of autoimmune lesions: Lessons from Sjogren’s syndrome (autoimmune epithelitis). Lupus 2006, 15, 255–261. [Google Scholar] [CrossRef] [PubMed]
- Katsiougiannis, S.; Tenta, R.; Skopouli, F.N. Autoimmune epithelitis (Sjogren’s syndrome); the impact of metabolic status of glandular epithelial cells on auto-immunogenicity. J. Autoimmun. 2019, 104, 102335. [Google Scholar] [CrossRef] [PubMed]
- Moustaka, K.; Katsiougiannis, S.; Tenta, R.; Havaki, S.; Koutsoudaki, P.; Moutsopoulos, H.M.; Skopouli, F. THU0223 Chronic adrenergic stimulation of minor salivary glands of patientswith primary sjögren’s drives er stress and activation of the unfolded protein response. Ann. Rheum. Dis. 2019, 78, 389. [Google Scholar] [CrossRef] [Green Version]
- B’Chir, W.; Maurin, A.C.; Carraro, V.; Averous, J.; Jousse, C.; Muranishi, Y.; Parry, L.; Stepien, G.; Fafournoux, P.; Bruhat, A. The eIF2alpha/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Res. 2013, 41, 7683–7699. [Google Scholar] [CrossRef] [Green Version]
- Deegan, S.; Saveljeva, S.; Gorman, A.M.; Samali, A. Stress-induced self-cannibalism: On the regulation of autophagy by endoplasmic reticulum stress. Cell. Mol. Life Sci. CMLS 2013, 70, 2425–2441. [Google Scholar] [CrossRef]
- Senft, D.; Ronai, Z.A. UPR, autophagy, and mitochondria crosstalk underlies the ER stress response. Trends Biochem. Sci. 2015, 40, 141–148. [Google Scholar] [CrossRef] [Green Version]
- Katsiougiannis, S.; Tenta, R.; Skopouli, F.N. Endoplasmic reticulum stress causes autophagy and apoptosis leading to cellular redistribution of the autoantigens Ro/Sjogren’s syndrome-related antigen A (SSA) and La/SSB in salivary gland epithelial cells. Clin. Exp. Immunol. 2015, 181, 244–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Obeid, M.; Tesniere, A.; Ghiringhelli, F.; Fimia, G.M.; Apetoh, L.; Perfettini, J.L.; Castedo, M.; Mignot, G.; Panaretakis, T.; Casares, N.; et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 2007, 13, 54–61. [Google Scholar] [CrossRef] [PubMed]
- Mavragani, C.P.; Crow, M.K. Activation of the type I interferon pathway in primary Sjogren’s syndrome. J. Autoimmun. 2010, 35, 225–231. [Google Scholar] [CrossRef] [PubMed]
- Crow, M.K. Long interspersed nuclear elements (LINE-1): Potential triggers of systemic autoimmune disease. Autoimmunity 2010, 43, 7–16. [Google Scholar] [CrossRef] [PubMed]
- Kalogirou, E.-M.; Piperi, E.P.; Tosios, K.I.; Tsiambas, E.; Fanourakis, G.; Sklavounou, A. Ductal cells of minor salivary glands in Sjögren’s syndrome express LINE-1 ORF2p and APOBEC3B. J. Oral. Pathol. Med. 2018, 47, 179–185. [Google Scholar] [CrossRef]
- Rodic, N.; Sharma, R.; Sharma, R.; Zampella, J.; Dai, L.; Taylor, M.S.; Hruban, R.H.; Iacobuzio-Donahue, C.A.; Maitra, A.; Torbenson, M.S.; et al. Long interspersed element-1 protein expression is a hallmark of many human cancers. Am. J. Pathol. 2014, 184, 1280–1286. [Google Scholar] [CrossRef] [Green Version]
- Carbone, A.; Gloghini, A.; Ferlito, A. 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. 2000, 109, 1170–1175. [Google Scholar] [CrossRef]
- Rischmueller, M.; Tieu, J.; Lester, S. Primary Sjogren’s syndrome. Best Pract. Res. Clin. Rheumatol. 2016, 30, 189–220. [Google Scholar] [CrossRef]
- Bartoloni, E.; Alunno, A.; Bistoni, O.; Caterbi, S.; Luccioli, F.; Santoboni, G.; Mirabelli, G.; Cannarile, F.; Gerli, R. Characterization of circulating endothelial microparticles and endothelial progenitor cells in primary Sjogren’s syndrome: New markers of chronic endothelial damage? Rheumatology 2015, 54, 536–544. [Google Scholar] [CrossRef] [Green Version]
- Mikulowska-Mennis, A.; Xu, B.; Berberian, J.M.; Michie, S.A. Lymphocyte migration to inflamed lacrimal glands is mediated by vascular cell adhesion molecule-1/alpha(4)beta(1) integrin, peripheral node addressin/l-selectin, and lymphocyte function-associated antigen-1 adhesion pathways. Am. J. Pathol. 2001, 159, 671–681. [Google Scholar] [CrossRef]
- Turkcapar, N.; Sak, S.D.; Saatci, M.; Duman, M.; Olmez, U. Vasculitis and expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin in salivary glands of patients with Sjogren’s syndrome. J. Rheumatol. 2005, 32, 1063–1070. [Google Scholar] [PubMed]
- Nagy, J.A.; Benjamin, L.; Zeng, H.; Dvorak, A.M.; Dvorak, H.F. Vascular permeability, vascular hyperpermeability and angiogenesis. Angiogenesis 2008, 11, 109–119. [Google Scholar] [CrossRef] [Green Version]
- Nayar, S.; Campos, J.; Chung, M.M.; Navarro-Nunez, L.; Chachlani, M.; Steinthal, N.; Gardner, D.H.; Rankin, P.; Cloake, T.; Caamano, J.H.; et al. 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. 2016, 197, 1957–1967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Margulies, D.H. The in-betweeners: MAIT cells join the innate-like lymphocytes gang. J. Exp. Med. 2014, 211, 1501–1502. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.; Gilfillan, S.; Kim, S.; Thompson, B.; Wang, X.; Sant, A.J.; Fremont, D.H.; Lantz, O.; Hansen, T.H. MR1 uses an endocytic pathway to activate mucosal-associated invariant T cells. J. Exp. Med. 2008, 205, 1201–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, S.; Martin, E.; Kim, S.; Yu, L.; Soudais, C.; Fremont, D.H.; Lantz, O.; Hansen, T.H. MR1 antigen presentation to mucosal-associated invariant T cells was highly conserved in evolution. Proc. Natl. Acad. Sci. USA 2009, 106, 8290–8295. [Google Scholar] [CrossRef] [Green Version]
- Matsui, Y.; Shapiro, H.M.; Sheehy, M.J.; Christenson, L.; Staunton, D.E.; Eynon, E.E.; Yunis, E.J. Differential expression of T cell differentiation antigens and major histocompatibility antigens on activated T cells during the cell cycle. Eur. J. Immunol. 1986, 16, 248–251. [Google Scholar] [CrossRef]
- Miles, J.J.; McCluskey, J.; Rossjohn, J.; Gras, S. Understanding the complexity and malleability of T-cell recognition. Immunol. Cell Biol. 2015, 93, 433–441. [Google Scholar] [CrossRef]
- Rossjohn, J.; Gras, S.; Miles, J.J.; Turner, S.J.; Godfrey, D.I.; McCluskey, J. T cell antigen receptor recognition of antigen-presenting molecules. Annu. Rev. Immunol. 2015, 33, 169–200. [Google Scholar] [CrossRef]
- Grimaldi, D.; Le Bourhis, L.; Sauneuf, B.; Dechartres, A.; Rousseau, C.; Ouaaz, F.; Milder, M.; Louis, D.; Chiche, J.D.; Mira, J.P.; et al. Specific MAIT cell behaviour among innate-like T lymphocytes in critically ill patients with severe infections. Intensive Care Med. 2014, 40, 192–201. [Google Scholar] [CrossRef]
- Ussher, J.E.; Bilton, M.; Attwod, E.; Shadwell, J.; Richardson, R.; de Lara, C.; Mettke, E.; Kurioka, A.; Hansen, T.H.; Klenerman, P.; et al. CD161++ CD8+ T cells, including the MAIT cell subset, are specifically activated by IL-12+IL-18 in a TCR-independent manner. Eur. J. Immunol. 2014, 44, 195–203. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Macardle, C.; Weedon, H.; Beroukas, D.; Banovic, T. Mucosal-associated invariant T cells are reduced and functionally immature in the peripheral blood of primary Sjögren’s syndrome patients. Eur. J. Immunol. 2016, 46, 2444–2453. [Google Scholar] [CrossRef]
- Fernandez, C.S.; Amarasena, T.; Kelleher, A.D.; Rossjohn, J.; McCluskey, J.; Godfrey, D.I.; Kent, S.J. MAIT cells are depleted early but retain functional cytokine expression in HIV infection. Immunol. Cell Biol. 2015, 93, 177–188. [Google Scholar] [CrossRef] [PubMed]
- Reantragoon, R.; Corbett, A.J.; Sakala, I.G.; Gherardin, N.A.; Furness, J.B.; Chen, Z.; Eckle, S.B.; Uldrich, A.P.; Birkinshaw, R.W.; Patel, O.; et al. Antigen-loaded MR1 tetramers define T cell receptor heterogeneity in mucosal-associated invariant T cells. J. Exp. Med. 2013, 210, 2305–2320. [Google Scholar] [CrossRef]
- Rusakiewicz, S.; Nocturne, G.; Lazure, T.; Semeraro, M.; Flament, C.; Caillat-Zucman, S.; Sene, D.; Delahaye, N.; Vivier, E.; Chaba, K.; et al. NCR3/NKp30 contributes to pathogenesis in primary Sjogren’s syndrome. Sci. Transl. Med. 2013, 5, 195ra196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Izumi, Y.; Ida, H.; Huang, M.; Iwanaga, N.; Tanaka, F.; Aratake, K.; Arima, K.; Tamai, M.; Kamachi, M.; Nakamura, H.; et al. Characterization of peripheral natural killer cells in primary Sjogren’s syndrome: Impaired NK cell activity and low NK cell number. J. Lab. Clin. Med. 2006, 147, 242–249. [Google Scholar] [CrossRef]
- Rizzo, C.; La Barbera, L.; Lo Pizzo, M.; Ciccia, F.; Sireci, G.; Guggino, G. Invariant NKT Cells and Rheumatic Disease: Focus on Primary Sjogren Syndrome. Int. J. Mol. Sci. 2019, 20, 5435. [Google Scholar] [CrossRef] [Green Version]
- Godfrey, D.I.; MacDonald, H.R.; Kronenberg, M.; Smyth, M.J.; Van Kaer, L. NKT cells: What’s in a name? Nat. Rev. Immunol. 2004, 4, 231–237. [Google Scholar] [CrossRef]
- Godfrey, D.I.; Kronenberg, M. Going both ways: Immune regulation via CD1d-dependent NKT cells. J. Clin. Investig. 2004, 114, 1379–1388. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.Q.; Wen, X.; Kim, P.J.; Singh, R.R. Invariant NKT cells inhibit autoreactive B cells in a contact- and CD1d-dependent manner. J. Immunol. 2011, 186, 1512–1520. [Google Scholar] [CrossRef] [Green Version]
- Sudzius, G.; Mieliauskaite, D.; Siaurys, A.; Viliene, R.; Butrimiene, I.; Characiejus, D.; Dumalakiene, I. Distribution of Peripheral Lymphocyte Populations in Primary Sjogren’s Syndrome Patients. J. Immunol. Res. 2015, 2015, 854706. [Google Scholar] [CrossRef] [PubMed]
- van der Vliet, H.J.; von Blomberg, B.M.; Nishi, N.; Reijm, M.; Voskuyl, A.E.; van Bodegraven, A.A.; Polman, C.H.; Rustemeyer, T.; Lips, P.; van den Eertwegh, A.J.; et al. Circulating V(alpha24+) Vbeta11+ NKT cell numbers are decreased in a wide variety of diseases that are characterized by autoreactive tissue damage. Clin. Immunol. 2001, 100, 144–148. [Google Scholar] [CrossRef] [PubMed]
- Guggino, G.; Ciccia, F.; Raimondo, S.; Giardina, G.; Alessandro, R.; Dieli, F.; Sireci, G.; Triolo, G. Invariant NKT cells are expanded in peripheral blood but are undetectable in salivary glands of patients with primary Sjogren’s syndrome. Clin. Exp. Rheumatol. 2016, 34, 25–31. [Google Scholar] [PubMed]
- Ahn, Y.O.; Blazar, B.R.; Miller, J.S.; Verneris, M.R. Lineage relationships of human interleukin-22-producing CD56+ RORgammat+ innate lymphoid cells and conventional natural killer cells. Blood 2013, 121, 2234–2243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciccia, F.; Guggino, G.; Rizzo, A.; Ferrante, A.; Raimondo, S.; Giardina, A.; Dieli, F.; Campisi, G.; Alessandro, R.; Triolo, G. Potential involvement of IL-22 and IL-22-producing cells in the inflamed salivary glands of patients with Sjogren’s syndrome. Ann. Rheum. Dis. 2012, 71, 295–301. [Google Scholar] [CrossRef]
- Mjosberg, J.; Eidsmo, L. Update on innate lymphoid cells in atopic and non-atopic inflammation in the airways and skin. Clin. Exp. Allergy J. Br. Soc. Allergy Clin. Immunol. 2014, 44, 1033–1043. [Google Scholar] [CrossRef]
- Shikhagaie, M.M.; Germar, K.; Bal, S.M.; Ros, X.R.; Spits, H. Innate lymphoid cells in autoimmunity: Emerging regulators in rheumatic diseases. Nat. Rev. Rheumatol. 2017, 13, 164–173. [Google Scholar] [CrossRef]
- Zook, E.C.; Kee, B.L. Development of innate lymphoid cells. Nat. Immunol. 2016, 17, 775–782. [Google Scholar] [CrossRef]
- Drake, L.Y.; Iijima, K.; Kita, H. Group 2 innate lymphoid cells and CD4+ T cells cooperate to mediate type 2 immune response in mice. Allergy 2014, 69, 1300–1307. [Google Scholar] [CrossRef] [Green Version]
- Ebbo, M.; Crinier, A.; Vely, F.; Vivier, E. Innate lymphoid cells: Major players in inflammatory diseases. Nat. Rev. Immunol. 2017, 17, 665–678. [Google Scholar] [CrossRef]
- Bernink, J.H.; Krabbendam, L.; Germar, K.; de Jong, E.; Gronke, K.; Kofoed-Nielsen, M.; Munneke, J.M.; Hazenberg, M.D.; Villaudy, J.; Buskens, C.J.; et al. Interleukin-12 and -23 Control Plasticity of CD127(+) Group 1 and Group 3 Innate Lymphoid Cells in the Intestinal Lamina Propria. Immunity 2015, 43, 146–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ohne, Y.; Silver, J.S.; Thompson-Snipes, L.; Collet, M.A.; Blanck, J.P.; Cantarel, B.L.; Copenhaver, A.M.; Humbles, A.A.; Liu, Y.J. IL-1 is a critical regulator of group 2 innate lymphoid cell function and plasticity. Nat. Immunol. 2016, 17, 646–655. [Google Scholar] [CrossRef] [PubMed]
- Klose, C.S.; Artis, D. Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat. Immunol. 2016, 17, 765–774. [Google Scholar] [CrossRef] [PubMed]
- Qiu, J.; Guo, X.; Chen, Z.M.; He, L.; Sonnenberg, G.F.; Artis, D.; Fu, Y.X.; Zhou, L. Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora. Immunity 2013, 39, 386–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bal, S.M.; Bernink, J.H.; Nagasawa, M.; Groot, J.; Shikhagaie, M.M.; Golebski, K.; van Drunen, C.M.; Lutter, R.; Jonkers, R.E.; Hombrink, P.; et al. IL-1beta, IL-4 and IL-12 control the fate of group 2 innate lymphoid cells in human airway inflammation in the lungs. Nat. Immunol. 2016, 17, 636–645. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Carrio, J.; Hahnlein, J.S.; Ramwadhdoebe, T.H.; Semmelink, J.F.; Choi, I.Y.; van Lienden, K.P.; Maas, M.; Gerlag, D.M.; Tak, P.P.; Geijtenbeek, T.B.; et al. Brief Report: Altered Innate Lymphoid Cell Subsets in Human Lymph Node Biopsy Specimens Obtained During the At-Risk and Earliest Phases of Rheumatoid Arthritis. Arthritis Rheumatol. 2017, 69, 70–76. [Google Scholar] [CrossRef]
- Leijten, E.F.; van Kempen, T.S.; Boes, M.; Michels-van Amelsfort, J.M.; Hijnen, D.; Hartgring, S.A.; van Roon, J.A.; Wenink, M.H.; Radstake, T.R. Brief report: Enrichment of activated group 3 innate lymphoid cells in psoriatic arthritis synovial fluid. Arthritis Rheumatol. 2015, 67, 2673–2678. [Google Scholar] [CrossRef] [Green Version]
- Ciccia, F.; Accardo-Palumbo, A.; Alessandro, R.; Rizzo, A.; Principe, S.; Peralta, S.; Raiata, F.; Giardina, A.; De Leo, G.; Triolo, G. Interleukin-22 and interleukin-22-producing NKp44+ natural killer cells in subclinical gut inflammation in ankylosing spondylitis. Arthritis Rheumatol. 2012, 64, 1869–1878. [Google Scholar] [CrossRef]
- Ciccia, F.; Guggino, G.; Rizzo, A.; Saieva, L.; Peralta, S.; Giardina, A.; Cannizzaro, A.; Sireci, G.; De Leo, G.; Alessandro, R.; et al. Type 3 innate lymphoid cells producing IL-17 and IL-22 are expanded in the gut, in the peripheral blood, synovial fluid and bone marrow of patients with ankylosing spondylitis. Ann. Rheumatol. Dis. 2015, 74, 1739–1747. [Google Scholar] [CrossRef]
- Brito-Zeron, P.; Baldini, C.; Bootsma, H.; Bowman, S.J.; Jonsson, R.; Mariette, X.; Sivils, K.; Theander, E.; Tzioufas, A.; Ramos-Casals, M. Sjogren syndrome. Nat. Rev. Dis. Primers 2016, 2, 16047. [Google Scholar] [CrossRef] [PubMed]
- Pan, H.F.; Li, X.P.; Zheng, S.G.; Ye, D.Q. Emerging role of interleukin-22 in autoimmune diseases. Cytokine Growth Factor Rev. 2013, 24, 51–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cella, M.; Fuchs, A.; Vermi, W.; Facchetti, F.; Otero, K.; Lennerz, J.K.; Doherty, J.M.; Mills, J.C.; Colonna, M. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 2009, 457, 722–725. [Google Scholar] [CrossRef] [PubMed]
- Hoorweg, K.; Peters, C.P.; Cornelissen, F.; Aparicio-Domingo, P.; Papazian, N.; Kazemier, G.; Mjosberg, J.M.; Spits, H.; Cupedo, T. Functional Differences between Human NKp44(-) and NKp44(+) RORC(+) Innate Lymphoid Cells. Front. Immunol. 2012, 3, 72. [Google Scholar] [CrossRef] [Green Version]
- Glatzer, T.; Killig, M.; Meisig, J.; Ommert, I.; Luetke-Eversloh, M.; Babic, M.; Paclik, D.; Bluthgen, N.; Seidl, R.; Seifarth, C.; et al. RORgammat(+) innate lymphoid cells acquire a proinflammatory program upon engagement of the activating receptor NKp44. Immunity 2013, 38, 1223–1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parodi, M.; Favoreel, H.; Candiano, G.; Gaggero, S.; Sivori, S.; Mingari, M.C.; Moretta, L.; Vitale, M.; Cantoni, C. NKp44-NKp44 Ligand Interactions in the Regulation of Natural Killer Cells and Other Innate Lymphoid Cells in Humans. Front. Immunol. 2019, 10, 719. [Google Scholar] [CrossRef] [PubMed]
- Lavoie, T.N.; Stewart, C.M.; Berg, K.M.; Li, Y.; Nguyen, C.Q. Expression of interleukin-22 in Sjogren’s syndrome: Significant correlation with disease parameters. Scand. J. Immunol. 2011, 74, 377–382. [Google Scholar] [CrossRef]
- Sonnenberg, G.F.; Nair, M.G.; Kirn, T.J.; Zaph, C.; Fouser, L.A.; Artis, D. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. J. Exp. Med. 2010, 207, 1293–1305. [Google Scholar] [CrossRef] [Green Version]
- Savan, R.; McFarland, A.P.; Reynolds, D.A.; Feigenbaum, L.; Ramakrishnan, K.; Karwan, M.; Shirota, H.; Klinman, D.M.; Dunleavy, K.; Pittaluga, S.; et al. A novel role for IL-22R1 as a driver of inflammation. Blood 2011, 117, 575–584. [Google Scholar] [CrossRef] [Green Version]
- Huber, S.; Gagliani, N.; Zenewicz, L.A.; Huber, F.J.; Bosurgi, L.; Hu, B.; Hedl, M.; Zhang, W.; O’Connor, W., Jr.; Murphy, A.J.; et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature 2012, 491, 259–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciccia, F.; Guggino, G.; Rizzo, A.; Bombardieri, M.; Raimondo, S.; Carubbi, F.; Cannizzaro, A.; Sireci, G.; Dieli, F.; Campisi, G.; et al. Interleukin (IL)-22 receptor 1 is over-expressed in primary Sjogren’s syndrome and Sjogren-associated non-Hodgkin lymphomas and is regulated by IL-18. Clin. Exp. Immunol. 2015, 181, 219–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cella, M.; Miller, H.; Song, C. Beyond NK cells: The expanding universe of innate lymphoid cells. Front. Immunol. 2014, 5, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Groom, J.; Kalled, S.L.; Cutler, A.H.; Olson, C.; Woodcock, S.A.; Schneider, P.; Tschopp, J.; Cachero, T.G.; Batten, M.; Wheway, J.; et al. Association of BAFF/BLyS overexpression and altered B cell differentiation with Sjogren’s syndrome. J. Clin. Investig. 2002, 109, 59–68. [Google Scholar] [CrossRef]
- Bar-Ephraim, Y.E.; Cornelissen, F.; Papazian, N.; Konijn, T.; Hoogenboezem, R.M.; Sanders, M.A.; Westerman, B.A.; Gonultas, M.; Kwekkeboom, J.; Den Haan, J.M.M.; et al. Cross-Tissue Transcriptomic Analysis of Human Secondary Lymphoid Organ-Residing ILC3s Reveals a Quiescent State in the Absence of Inflammation. Cell Rep. 2017, 21, 823–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wenink, M.H.; Leijten, E.F.A.; Cupedo, T.; Radstake, T. Review: Innate Lymphoid Cells: Sparking Inflammatory Rheumatic Disease? Arthritis Rheumatol. 2017, 69, 885–897. [Google Scholar] [CrossRef] [PubMed]
- Bodewes, I.L.A.; Bjork, A.; Versnel, M.A.; Wahren-Herlenius, M. Innate immunity and interferons in the pathogenesis of Sjogren’s syndrome. Rheumatology 2019. [Google Scholar] [CrossRef]
- Davies, R.; Sarkar, I.; Hammenfors, D.; Bergum, B.; Vogelsang, P.; Solberg, S.M.; Gavasso, S.; Brun, J.G.; Jonsson, R.; Appel, S. Single Cell Based Phosphorylation Profiling Identifies Alterations in Toll-Like Receptor 7 and 9 Signaling in Patients With Primary Sjogren’s Syndrome. Front. Immunol. 2019, 10, 281. [Google Scholar] [CrossRef] [PubMed]
- Schoenborn, J.R.; Wilson, C.B. Regulation of interferon-gamma during innate and adaptive immune responses. Adv. Immunol. 2007, 96, 41–101. [Google Scholar] [CrossRef]
- Bodewes, I.L.A.; Al-Ali, S.; van Helden-Meeuwsen, C.G.; Maria, N.I.; Tarn, J.; Lendrem, D.W.; Schreurs, M.W.J.; Steenwijk, E.C.; van Daele, P.L.A.; Both, T.; et al. Systemic interferon type I and type II signatures in primary Sjogren’s syndrome reveal differences in biological disease activity. Rheumatology 2018, 57, 921–930. [Google Scholar] [CrossRef] [Green Version]
- Donnelly, R.P.; Kotenko, S.V. Interferon-lambda: A new addition to an old family. J. Interferon Cytokine Res. 2010, 30, 555–564. [Google Scholar] [CrossRef] [Green Version]
- Ronnblom, L. The importance of the type I interferon system in autoimmunity. Clin. Exp. Rheumatol. 2016, 34, 21–24. [Google Scholar]
- Fitzgerald-Bocarsly, P.; Dai, J.; Singh, S. Plasmacytoid dendritic cells and type I IFN: 50 years of convergent history. Cytokine Growth Factor Rev. 2008, 19, 3–19. [Google Scholar] [CrossRef] [Green Version]
- Maria, N.I.; Steenwijk, E.C.; AS, I.J.; van Helden-Meeuwsen, C.G.; Vogelsang, P.; Beumer, W.; Brkic, Z.; van Daele, P.L.; van Hagen, P.M.; van der Spek, P.J.; et al. Contrasting expression pattern of RNA-sensing receptors TLR7, RIG-I and MDA5 in interferon-positive and interferon-negative patients with primary Sjogren’s syndrome. Ann. Rheum. Dis. 2017, 76, 721–730. [Google Scholar] [CrossRef] [PubMed]
- Barizzone, N.; Monti, S.; Mellone, S.; Godi, M.; Marchini, M.; Scorza, R.; Danieli, M.G.; D’Alfonso, S. Rare Variants in the TREX1 Gene and Susceptibility to Autoimmune Diseases. Biomed. Res. Int. 2013, 2013, 471703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vlachogiannis, N.I.; Nezos, A.; Tzioufas, A.G.; Koutsilieris, M.; Moutsopoulos, H.M.; Mavragani, C.P. Increased frequency of the PTPN22W* variant in primary Sjogren’s Syndrome: Association with low type I IFN scores. Clin. Immunol. 2016, 173, 157–160. [Google Scholar] [CrossRef]
- Piccinini, A.M.; Midwood, K.S. DAMPening inflammation by modulating TLR signalling. Mediat. Inflamm. 2010, 2010, 672395. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Cao, X. Cellular and molecular regulation of innate inflammatory responses. Cell Mol. Immunol. 2016, 13, 711–721. [Google Scholar] [CrossRef]
- Akira, S.; Takeda, K. Toll-like receptor signalling. Nat. Rev. Immunol 2004, 4, 499–511. [Google Scholar] [CrossRef]
- Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spachidou, M.P.; Bourazopoulou, E.; Maratheftis, C.I.; Kapsogeorgou, E.K.; Moutsopoulos, H.M.; Tzioufas, A.G.; Manoussakis, M.N. Expression of functional Toll-like receptors by salivary gland epithelial cells: Increased mRNA expression in cells derived from patients with primary Sjogren’s syndrome. Clin. Exp. Immunol. 2007, 147, 497–503. [Google Scholar] [CrossRef]
- Kiripolsky, J.; McCabe, L.G.; Gaile, D.P.; Kramer, J.M. Myd88 is required for disease development in a primary Sjogren’s syndrome mouse model. J. Leukoc. Biol. 2017, 102, 1411–1420. [Google Scholar] [CrossRef] [Green Version]
- Shimizu, T.; Nakamura, H.; Takatani, A.; Umeda, M.; Horai, Y.; Kurushima, S.; Michitsuji, T.; Nakashima, Y.; Kawakami, A. Activation of Toll-like receptor 7 signaling in labial salivary glands of primary Sjogren’s syndrome patients. Clin. Exp. Immunol. 2019, 196, 39–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, L.; Zhang, Z.; Yu, C.; Yang, C. Expression of Toll-like receptors 7, 8, and 9 in primary Sjogren’s syndrome. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2010, 109, 844–850. [Google Scholar] [CrossRef] [PubMed]
- Shimizu, S.; Kurashige, Y.; Nishimura, M.; Mami, Y.; Sato, J.; Saitoh, M.; Selimović, D.; Abiko, Y. Involvement of toll-like receptors in autoimmune sialoadenitis of the non-obese diabetic mouse. J. Oral Pathol. Med. 2012, 41, 517–523. [Google Scholar] [CrossRef] [PubMed]
- Kwok, S.K.; Cho, M.L.; Her, Y.M.; Oh, H.J.; Park, M.K.; Lee, S.Y.; Woo, Y.J.; Ju, J.H.; Park, K.S.; Kim, H.Y.; et al. TLR2 ligation induces the production of IL-23/IL-17 via IL-6, STAT3 and NF-kB pathway in patients with primary Sjogren’s syndrome. Arthritis Res. 2012, 14, R64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ittah, M.; Miceli-Richard, C.; Gottenberg, J.E.; Sellam, J.; Eid, P.; Lebon, P.; Pallier, C.; Lepajolec, C.; Mariette, X. Viruses induce high expression of BAFF by salivary gland epithelial cells through TLR- and type-I IFN-dependent and -independent pathways. Eur. J. Immunol. 2008, 38, 1058–1064. [Google Scholar] [CrossRef]
- Bombardieri, M.; Barone, F.; Pittoni, V.; Alessandri, C.; Conigliaro, P.; Blades, M.C.; Priori, R.; McInnes, I.B.; Valesini, G.; Pitzalis, C. Increased circulating levels and salivary gland expression of interleukin-18 in patients with Sjogren’s syndrome: Relationship with autoantibody production and lymphoid organization of the periductal inflammatory infiltrate. Arthritis Res. 2004, 6, R447–R456. [Google Scholar] [CrossRef] [Green Version]
- Kiefer, K.; Oropallo, M.A.; Cancro, M.P.; Marshak-Rothstein, A. Role of type I interferons in the activation of autoreactive B cells. Immunol. Cell Biol. 2012, 90, 498–504. [Google Scholar] [CrossRef] [Green Version]
- Charras, A.; Arvaniti, P.; Le Dantec, C.; Dalekos, G.N.; Zachou, K.; Bordron, A.; Renaudineau, Y. JAK Inhibitors and Oxidative Stress Control. Front. Immunol. 2019, 10, 2814. [Google Scholar] [CrossRef]
- Baldini, C.; Rossi, C.; Ferro, F.; Santini, E.; Seccia, V.; Donati, V.; Solini, A. The P2X7 receptor-inflammasome complex has a role in modulating the inflammatory response in primary Sjogren’s syndrome. J. Intern. Med. 2013, 274, 480–489. [Google Scholar] [CrossRef]
- Broz, P.; Dixit, V.M. Inflammasomes: Mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 2016, 16, 407–420. [Google Scholar] [CrossRef]
- Mariathasan, S.; Newton, K.; Monack, D.M.; Vucic, D.; French, D.M.; Lee, W.P.; Roose-Girma, M.; Erickson, S.; Dixit, V.M. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 2004, 430, 213–218. [Google Scholar] [CrossRef] [PubMed]
- Winsor, N.; Krustev, C.; Bruce, J.; Philpott, D.J.; Girardin, S.E. Canonical and noncanonical inflammasomes in intestinal epithelial cells. Cell. Microbiol. 2019, 21, e13079. [Google Scholar] [CrossRef] [PubMed]
- Woods, L.T.; Camden, J.M.; Batek, J.M.; Petris, M.J.; Erb, L.; Weisman, G.A. P2X7 receptor activation induces inflammatory responses in salivary gland epithelium. Am. J. Physiol. Cell Physiol. 2012, 303, C790–C801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imgenberg-Kreuz, J.; Sandling, J.K.; Nordmark, G. Epigenetic alterations in primary Sjogren’s syndrome—An overview. Clin. Immunol. 2018, 196, 12–20. [Google Scholar] [CrossRef] [PubMed]
- Lessard, C.J.; Li, H.; Adrianto, I.; Ice, J.A.; Rasmussen, A.; Grundahl, K.M.; Kelly, J.A.; Dozmorov, M.G.; Miceli-Richard, C.; Bowman, S.; et al. Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjogren’s syndrome. Nat. Genet. 2013, 45, 1284–1292. [Google Scholar] [CrossRef] [PubMed]
- Holoch, D.; Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat. Rev. Genet. 2015, 16, 71–84. [Google Scholar] [CrossRef] [PubMed]
- Wu, G.C.; Pan, H.F.; Leng, R.X.; Wang, D.G.; Li, X.P.; Li, X.M.; Ye, D.Q. Emerging role of long noncoding RNAs in autoimmune diseases. Autoimmun. Rev. 2015, 14, 798–805. [Google Scholar] [CrossRef]
- Zilahi, E.; Tarr, T.; Papp, G.; Griger, Z.; Sipka, S.; Zeher, M. Increased microRNA-146a/b, TRAF6 gene and decreased IRAK1 gene expressions in the peripheral mononuclear cells of patients with Sjogren’s syndrome. Immunol. Lett. 2012, 141, 165–168. [Google Scholar] [CrossRef]
- Shi, H.; Zheng, L.-y.; Zhang, P.; Yu, C.-q. miR-146a and miR-155 expression in PBMCs from patients with Sjögren’s syndrome. J. Oral Pathol. Med. 2014, 43, 792–797. [Google Scholar] [CrossRef]
- Gourzi, V.C.; Kapsogeorgou, E.K.; Kyriakidis, N.C.; Tzioufas, A.G. Study of microRNAs (miRNAs) that are predicted to target the autoantigens Ro/SSA and La/SSB in primary Sjögren’s Syndrome. Clin. Exp. Immunol. 2015, 182, 14–22. [Google Scholar] [CrossRef] [Green Version]
- Shi, H.; Cao, N.; Pu, Y.; Xie, L.; Zheng, L.; Yu, C. Long non-coding RNA expression profile in minor salivary gland of primary Sjögren’s syndrome. Arthritis Res. Ther. 2016, 18, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalunian, K.C.; Merrill, J.T.; Maciuca, R.; McBride, J.M.; Townsend, M.J.; Wei, X.; Davis, J.C., Jr.; Kennedy, W.P. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-alpha) in patients with systemic lupus erythematosus (ROSE). Ann. Rheum. Dis. 2016, 75, 196–202. [Google Scholar] [CrossRef] [PubMed]
- Khamashta, M.; Merrill, J.T.; Werth, V.P.; Furie, R.; Kalunian, K.; Illei, G.G.; Drappa, J.; Wang, L.; Greth, W. Sifalimumab, an anti-interferon-alpha monoclonal antibody, in moderate to severe systemic lupus erythematosus: A randomised, double-blind, placebo-controlled study. Ann. Rheum. Dis. 2016, 75, 1909–1916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Furie, R.; Khamashta, M.; Merrill, J.T.; Werth, V.P.; Kalunian, K.; Brohawn, P.; Illei, G.G.; Drappa, J.; Wang, L.; Yoo, S. Anifrolumab, an Anti-Interferon-alpha Receptor Monoclonal Antibody, in Moderate-to-Severe Systemic Lupus Erythematosus. Arthritis Rheumatol. 2017, 69, 376–386. [Google Scholar] [CrossRef] [Green Version]
- Burge, D.J.; Eisenman, J.; Byrnes-Blake, K.; Smolak, P.; Lau, K.; Cohen, S.B.; Kivitz, A.J.; Levin, R.; Martin, R.W.; Sherrer, Y.; et al. Safety, pharmacokinetics, and pharmacodynamics of RSLV-132, an RNase-Fc fusion protein in systemic lupus erythematosus: A randomized, double-blind, placebo-controlled study. Lupus 2017, 26, 825–834. [Google Scholar] [CrossRef]
- Lood, C.; Allhorn, M.; Lood, R.; Gullstrand, B.; Olin, A.I.; Ronnblom, L.; Truedsson, L.; Collin, M.; Bengtsson, A.A. IgG glycan hydrolysis by endoglycosidase S diminishes the proinflammatory properties of immune complexes from patients with systemic lupus erythematosus: A possible new treatment? Arthritis Rheum. 2012, 64, 2698–2706. [Google Scholar] [CrossRef]
- Guiducci, C.; Ghirelli, C.; Marloie-Provost, M.A.; Matray, T.; Coffman, R.L.; Liu, Y.J.; Barrat, F.J.; Soumelis, V. PI3K is critical for the nuclear translocation of IRF-7 and type I IFN production by human plasmacytoid predendritic cells in response to TLR activation. J. Exp. Med. 2008, 205, 315–322. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.W.; Tang, W.; Zuo, J.P. Toll-like receptors: Potential targets for lupus treatment. Acta Pharm. Sin. 2015, 36, 1395–1407. [Google Scholar] [CrossRef]
- Muskardin, T.L.W.; Niewold, T.B. Type I interferon in rheumatic diseases. Nat. Rev. Rheumatol. 2018, 14, 214–228. [Google Scholar] [CrossRef]
- Sanchez, G.A.M.; Reinhardt, A.; Ramsey, S.; Wittkowski, H.; Hashkes, P.J.; Berkun, Y.; Schalm, S.; Murias, S.; Dare, J.A.; Brown, D.; et al. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J. Clin. Investig. 2018, 128, 3041–3052. [Google Scholar] [CrossRef] [Green Version]
- Retamozo, S.; Flores-Chavez, A.; Consuegra-Fernandez, M.; Lozano, F.; Ramos-Casals, M.; Brito-Zeron, P. Cytokines as therapeutic targets in primary Sjogren syndrome. Pharmacol. Ther. 2018, 184, 81–97. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.W.; Zhou, P.R.; Wei, P.; Cong, X.; Wu, L.L.; Hua, H. Expression of interleukin-17 in primary Sjogren’s syndrome and the correlation with disease severity: A systematic review and meta-analysis. Scand. J. Immunol. 2018, 87, e12649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Devauchelle-Pensec, V.; Mariette, X.; Jousse-Joulin, S.; Berthelot, J.M.; Perdriger, A.; Puechal, X.; Le Guern, V.; Sibilia, J.; Gottenberg, J.E.; Chiche, L.; et al. Treatment of primary Sjogren syndrome with rituximab: A randomized trial. Ann. Intern. Med. 2014, 160, 233–242. [Google Scholar] [CrossRef] [PubMed]
- Kuznik, A.; Bencina, M.; Svajger, U.; Jeras, M.; Rozman, B.; Jerala, R. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J. Immunol. 2011, 186, 4794–4804. [Google Scholar] [CrossRef] [Green Version]
- Yoon, C.H.; Lee, H.J.; Lee, E.Y.; Lee, E.B.; Lee, W.-W.; Kim, M.K.; Wee, W.R. Effect of Hydroxychloroquine Treatment on Dry Eyes in Subjects with Primary Sjögren’s Syndrome: A Double-Blind Randomized Control Study. J. Korean Med. Sci. 2016, 31, 1127–1135. [Google Scholar] [CrossRef]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Rizzo, C.; Grasso, G.; Destro Castaniti, G.M.; Ciccia, F.; Guggino, G. Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation. Vaccines 2020, 8, 272. https://doi.org/10.3390/vaccines8020272
Rizzo C, Grasso G, Destro Castaniti GM, Ciccia F, Guggino G. Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation. Vaccines. 2020; 8(2):272. https://doi.org/10.3390/vaccines8020272
Chicago/Turabian StyleRizzo, Chiara, Giulia Grasso, Giulia Maria Destro Castaniti, Francesco Ciccia, and Giuliana Guggino. 2020. "Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation" Vaccines 8, no. 2: 272. https://doi.org/10.3390/vaccines8020272
APA StyleRizzo, C., Grasso, G., Destro Castaniti, G. M., Ciccia, F., & Guggino, G. (2020). Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation. Vaccines, 8(2), 272. https://doi.org/10.3390/vaccines8020272