References
1. Lisnevskaia L, Murphy G, Isenberg D. Systemic lupus erythematosus.Lancet 2014; 384 :1878-88.
2. Zharkova O, Celhar T, Cravens PD, Satterthwaite AB, Fairhurst AM, Davis LS. Pathways leading to an immunological disease: systemic lupus erythematosus. Rheumatology 2017; 56 :i55-66.
3. You M, Dong G, Li F, Ma F, Ren J, Xu Y, Yue H, Tang R, Ren D, Hou Y. Ligation of CD180 inhibits IFN-α signaling in a Lyn-PI3K-BTK-dependent manner in B cells. Cell Mol Immunol 2017; 14 :192-202.
4. Dörner T, Giesecke C, Lipsky PE. Mechanisms of B cell autoimmunity in SLE. Arthritis Res Ther 2011; 13 :243.
5. Kotzin BL. Systemic lupus erythematosus. Cell 1996;85 :303.
6. Alexander JJ, Jacob A, Chang A, Quigg RJ, Jarvis JN. Double negative T cells, a potential biomarker for systemic lupus erythematosus.Precision Clin Med 2020; 3 :34-43.
7. Chesnutt MS, Finck BK, Killeen N, Connolly MK, Goodman H, Wofsy D. Enhanced lymphoproliferation and diminished autoimmunity in CD4-deficient MRL/lpr mice. Clin Immunol Immunopathol 1998;87 :23-32.
8. Nagasu A, Mukai T, Iseki M, Kawahara K, Tsuji S, Nagasu H, Ueki Y, Ishihara K, Kashihara N, Morita Y. Sh3bp2 Gain-of-function mutation ameliorates lupus phenotypes in B6.MR-Faslpr mice.Cells 2019; 8 :402.
9. Brandt D, Hedrich CM. TCRαβ + CD3 + CD4 CD8 (double negative) T cells in autoimmunity. Autoimmun Rev 2018; 17 :422-30.
10. Martina MN, Noel S, Saxena A, Rabb H, Hamad AR. Double negative (DN) αβ T cells: misperception and overdue recognition. Immunol Cell Biol 2015; 93 :305.
11. Shaltout AS, Sayed D, Badary MS, Nafee AM, El Zohri MH, Bakry R, Ahmed SH. Effect of IL6 and IL23 on double negative T cells and anti ds-DNA in systemic lupus erythematosus patients. Hum Immunol2016; 77 :937-43.
12. Seagal J, Leider N, Wildbaum G, Karin N, Melamed D. Increased plasma cell frequency and accumulation of abnormal syndecan-1plus T-cells in Igmu-deficient/lpr mice. Int Immunol 2003; 15 :1045-52.
13. Mohamood AS, Bargatze D, Xiao Z, Jie C, Yagita H, Ruben D, Watson J, Chakravarti S, Schneck JP, Hamad AR. Fas-mediated apoptosis regulates the composition of peripheral αβ T cell repertoire by constitutively purging out double negative T cells. Plos One 2008;3 :e3465.
14. Liu L, Takeda K, Akkoyunlu M. Disease stage-specific pathogenicity of CD138 (Syndecan 1)-expressing T Cells in systemic lupus erythematosus. Front Immunol 2020; 11 :01569.
15. Couchman JR. Syndecans: proteoglycan regulators of cell-surface microdomains? Nature Rev Mol Cell Biol 2003; 4 :926-37.
16 Lu LD, Stump KL, Wallace NH, Dobrzanski P, Serdikoff C, Gingrich DE, Dugan BJ, Angeles TS, Albom MS, Mason JL, Ator MA, Dorsey BD, Ruggeri BA, Seavey MM. Depletion of autoreactive plasma cells and treatment of lupus nephritis in mice using CEP-33779, a novel, orally active, selective inhibitor of JAK2. J Immunol 2011;187 :3840-53.
17. Calame KL. Plasma cells: finding new light at the end of B cell development. Nat Immunol 2001; 2 :1103-08.
18. Pan Z, Chen M, Zhang Q, et al. CD3-positive plasmablastic B-cell neoplasms: a diagnostic pitfall. Mod Pathol 2018;31 :718-31.
19. Getachew Y, Cusimano FA, James LP, Thiele DL. The role of intrahepatic CD3 +/CD4 /CD8 double negative T (DN T) cells in enhanced acetaminophen toxicity. Toxicol Appl Pharmacol 2014;280 :264-71.
20. Benihoud K, Bonardelle D, Bobé P, Kiger N. MRL/lpr CD4- CD8- and CD8+ T cells, respectively, mediate Fas-dependent and perforin cytotoxic pathways. Eur J Immunol 1997; 27 :415-20.
21. Hidalgo Y, Núñez S, Fuenzalida MJ, Flores-Santibáñez F, Sáez PJ, Dorner J, Lennon-Dumenil AM, Martínez V, Zorn E, Rosemblatt M, Sauma D, Bono MR. Thymic B cells promote germinal center-like structures and the expansion of follicular helper T cells in lupus-prone mice. Front Immunol 2020; 11 :696.
22. Menon M, Blair PA, Isenberg DA, Mauri C. A regulatory feedback between plasmacytoid dendritic cells and regulatory B cells is aberrant in systemic lupus erythematosus. Immunity 2016;44 :683-97.
23. Liu J, Huang X, Hao S, Wang Y, Liu M, Xu J, Zhang X, Yu T, Gan S, Dai D, Luo X, Lu Q, Mao C, Zhang Y, Shen N, Li B, Huang M, Zhu X, Jin J, Cheng X, Sun SC, Xiao Y. Peli1 negatively regulates noncanonical NF-κB signaling to restrain systemic lupus erythematosus. Nat Commun2018; 9 : 1136.
24. Dik WA, Pike-Overzet K, Weerkamp F, de Ridder D, de Haas EF, Baert MR, van der Spek P, Koster EE, Reinders MJ, van Dongen JJ, Langerak AW, Staal FJ. New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling.J Exp Med 2005; 201 :1715-23.
25. Anderson G, Jenkinson EJ. Lymphostromal interactions in thymic development and function. Nat Rev Immunol 2001; 1 :31.
26. Trimble LA, Prince KA, Pestano GA, Daley J, Cantor H. Fas-dependent elimination of nonselected CD8 cells and lpr disease. J Immunol2002; 168 :4960-7.
27. Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA, Nagata S. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature 1992; 356 :314-7.
28. Suda T, Takahashi T, Golstein P, Nagata S. Molecular cloning and expression of the fas ligand, a novel member of the tumor necrosis factor family. Cell 1993; 75 :1169-78.
29. Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2016; 12 :716-30.
30. Laurent L, Le Fur A, Bloas RL, Néel M, Mary C, Moreau A, Poirier N, Vanhove B, Fakhouri F. Prevention of lupus nephritis development in NZB/NZW mice by selective blockade of CD28. Eur J Immunol 2017;47 :1368-76.
31. Corneth OBJ, Schaper F, Luk F, Asmawidjaja PS, Mus AMC, Horst G, Heeringa P, Hendriks RW, Westra J, Lubberts E. Lack of IL-17 receptor A signaling aggravates lymphoproliferation in C57BL/6 lpr mice. Sci Rep 2019; 9 :4032.
32. Zhou T, Bluethmann H, Eldridge J, Berry K, Mountz JD. Origin of CD4-CD8-B220+ T cells in MRL-lpr/lpr mice. Clues from a T cell receptor beta transgenic mouse. J Immunol 1993; 150 :3651-67.
33. Shirai T, Abe M, Yagita H, Okumura K, Morse HC. The expanded populations of CD4-CD8- T cell receptor alpha/beta+ T cells associated with the lpr and gld mutations are CD2. J Immunol 1990;144 :3756-61.
34. Giese T, Allison JP, Davidson WF. Functionally anergic lpr and gld B220+ T cell receptor (TCR)-alpha/beta+ double-negative T cells express CD28 and respond to costimulation with phorbol myristate acetate and antibodies to CD28 and the TCR. J Immunol 1993;151 :597-609.
35. Merino R, Fossati L, Iwamoto M, Takahashi S, Lemoine R, Ibnou-Zekri N, Pugliatti L, Merino J, Izui S. Effect of long-term anti-CD4 or anti-CD8 treatment on the development of lpr CD4- CD8- double negative T cells and of the autoimmune syndrome in MRL-lpr/lpr mice. J Autoimmun 1995; 8 :33-45.
36. Ohteki T, Iwamoto M, Izui S, MacDonald HR. Reduced development of CD4-8-B220+ T cells but normal autoantibody production in lpr/lpr mice lacking major histocompatibility complex class I molecules. Eur J Immunol 1995; 25 :37-41.
37. Crispín JC, Oukka M, Bayliss G, Cohen RA, Van Beek CA, Stillman IE, Kyttaris VC, Juang YT, Tsokos GC. Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol 2008; 181 :8761-6.
38. Grishkan IV, Ntranos A, Calabresi PA, Gocke AR. Helper T cells down-regulate CD4 expression upon chronic stimulation giving rise to double-negative T cells. Cell Immunol 2013; 284 :68-74.
39 Zhang D, Yang W, Degauque N, Tian Y, Mikita A, Zheng XX. New differentiation pathway for double-negative regulatory T cells that regulates the magnitude of immune responses. Blood 2007;109 :4071-9.
40. Hedrich CM, Rauen T, Crispin JC, Koga T, Ioannidis C, Zajdel M, Kyttaris VC, Tsokos GC. cAMP-responsive element modulator α (CREMα) trans-represses the transmembrane glycoprotein CD8 and contributes to the generation of CD3+CD4-CD8- T cells in health and disease. J Biol Chem 2013; 288 :31880-7.
41. Hedrich CM, Crispín JC, Rauen T, Ioannidis C, Koga T, Rodriguez Rodriguez N, Apostolidis SA, Kyttaris VC, Tsokos GC. cAMP responsive element modulator (CREM) α mediates chromatin remodeling of CD8 during the generation of CD3+ CD4- CD8- T cells. J Biol Chem 2014;289 :2361-70.
42. Li H, Adamopoulos IE, Moulton VR, et al. Systemic lupus erythematosus favors the generation of IL-17 producing double negative T cells. Nat Commun 2020; 11 :2859.
43. Sandhu V, Quan M. SLE and serum complement: causative, concomitant or coincidental? Open Rheumatol J 2017; 11 :113-22.
44. Tshilela KA, Ikeuchi H, Matsumoto T, Kuroiwa T, Sakurai N, Sakairi T, Kaneko Y, Maeshima A, Hiromura K, Nojima Y. Glomerular cytokine expression in murine lupus nephritis. Clin Exp Nephrol 2016;20 :23-9.
45. Selvaraja M, Abdullah M, Arip M, Chin VK, Shah A, Amin Nordin S. Elevated interleukin-25 and its association to Th2 cytokines in systemic lupus erythematosus with lupus nephritis. PloS One 2019;14 :e0224707.
46. Ehrenfeld M, Blank M, Shoenfeld Y, Hidvegi M. AVEMAR administration interferes with the Th2 response in experimental SLE and promotes amelioration of the disease. Lupus 2001; 10 :622-7.
47. Tan W, Gu Z, Leng J, Zou X, Chen H, Min F, Zhou W, Zhang L, Li G. Let-7f-5p ameliorates inflammation by targeting NLRP3 in bone marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Biomed Pharmacother 2019; 118 :109313.
48. Balomenos D, Rumold R, Theofilopoulos AN. Interferon-gamma is required for lupus-like disease and lymphoaccumulation in MRL-lpr mice.J Clin Invest 1998; 101 :364-71.
49. Juvet SC, Han M, Vanama R, Joe B, Kim EY, Zhao FL, Jeon C, Adeyi O, Zhang L. Autocrine IFNɣ controls the regulatory function of lymphoproliferative double negative T cells. PLoS One 2012;7 :e47732.
50. Tang X, Li W, Wen X, Zhang Z, Chen W, Yao G, Chen H, Wang D, Shi S, Sun L. Transplantation of dental tissue-derived mesenchymal stem cells ameliorates nephritis in lupus mice. Ann Transl Med 2019;7 :132.
51. Chodisetti SB, Fike AJ, Domeier PP, Singh H, Choi NM, Corradetti C, Kawasawa YI, Cooper TK, Caricchio R, Rahman ZSM. Type II but not type I IFN signaling is indispensable for TLR7-promoted development of autoreactive B cells and systemic autoimmunity. J Immunol 2020;204 :796-809.
52. Zhang X, Deriaud E, Jiao X, Braun D, Leclerc C, Lo-Man R. Type I interferons protect neonates from acute inflammation through interleukin 10-producing B cells. J Exp Med 2007; 204 :1107-18.
53. de la Varga-Martínez R, Rodríguez-Bayona B, Campos-Caro A, Añez GA, Medina-Varo F, Rodríguez C. Autoreactive B-lymphocytes in SLE and RA patients: isolation and characterisation using extractable nuclear and citrullinated antigens bound to immunobeads. Eur J Immunol 2019;49 :1107-16.
54 . Jackson SW, Davidson A. J BAFF inhibition in SLE—is tolerance restored? Immunol Rev 2019; 292 :102-19.
55. Mihaylova N, Chipinski P, Bradyanova S, Velikova T, Ivanova-Todorova E, Chausheva S, Herbáth M, Kalinova D, Prechl J, Kyurkchiev D, Tchorbanov AI. Suppression of autoreactive T and B lymphocytes by anti-annexin A1 antibody in a humanized NSG murine model of systemic lupus erythematosus. Clin Exp Immunol 2020; 199 :278-93.
56. Monroe JG, Bannish G, Fuentes-Panana EM, King LB, Sandel PC, Chung J, Sater R. Positive and negative selection during B lymphocyte development. Immunol Res 2003; 27 :427-42.
57. Xu X, Deobagkar-Lele M, Bull, KR, Crockford TL, Mead AJ, Cribbs AP, Sims D, Anzilotti C, Cornall RJ. An ontogenetic switch drives the positive and negative selection of B cells. Proc Nat Acad Sci2020; 117 :3718-27.
58. Benhamou D, Labi V, Novak R, Dai I, Shafir-Alon S, Weiss A, Gaujoux R, Arnold R, Shen-Orr SS, Rajewsky K, Melamed D. A c-Myc/miR17-92/Pten axis controls PI3K-mediated positive and negative selection in B cell development and reconstitutes CD19 deficiency. Cell Rep 2016;16 :419-31.
59. Akagi T, Yoshino T, Kondo E. The Fas antigen and Fas-mediated apoptosis in B-Cell differentiation. Leuk Lymphoma 1998;28 :483-89.
60. Hancz A, Koncz G, Szili D, Sármay G. TLR9-mediated signals rescue B-cells from Fas-induced apoptosis via inactivation of caspases.Immunol Lett 2012; 143 :77-84.
61. Akiyama C, Tsumiyama K, Uchimura C, Honda E, Miyazaki Y, Sakurai K, Miura Y, Hashiramoto A, Felsher DW, Shiozawa S. Conditional upregulation of IFN-α alone is sufficient to induce systemic lupus erythematosus.J Immunol 2019; 203 :835-43.
62. Giordani L, Sanchez M, Libri I, Quaranta MG, Mattioli B, Viora M. IFN-alpha amplifies human naive B cell TLR-9-mediated activation and Ig production. J Leukocyte Biol 2009; 86 :261.
63. Zheng N, Wang B, Fan J, Luo N, Kong Q, Ye H, Zhang J, Ming H, Yu X. Increased abundance of plasmacytoid dendritic cells and interferon-alpha induces plasma cell differentiation in patients of IgA nephropathy.Mediators Inflammation 2017; 2017 :1-15.
64. Jiang J, Zhao M, Chang C, Wu H, Lu Q. Type I interferons in the pathogenesis and treatment of autoimmune diseases. Clin Rev Allerg Immunol 2020; 59 (2):248-72.
65. Zeng J, Meng X, Zhou P, Yin Z, Xie Q, Zou H, Shen N, Ye Z, Tang Y. Interferon-α exacerbates neuropsychiatric phenotypes in lupus-prone mice. Arthritis Res Ther 2019; 21 :205.
66. Blanco P, Palucka AK, Gill M, Pascual V, Banchereau J. Induction of dendritic cell differentiation by IFN-α in systemic lupus erythematosus.Science 2001; 294 :1540-3.
67. Hron JD, Peng SL. Type I IFN protects against murine lupus. J Immunol 2004; 173 : 2134-42.
68. Wu Y, He S, Bai B, Zhang L, Xue L, Lin Z, Yang X, Zhu F, He P, Tang W, Zuo J. Therapeutic effects of the artemisinin analog SM934 on lupus-prone MRL/ lpr mice via inhibition of TLR-triggered B-cell activation and plasma cell formation. Cell Mol Immunol 2016;13 :379-90.
69. Nguyen KB, Cousens LP, Doughty LA, Pien GC, Durbin JE, Biron CA. Interferon α/β-mediated inhibition and promotion of interferon γ: STAT1 resolves a paradox. Nat Immunol 2000; 1 :70-6.
70. McKenna K, Beignon AS, Bhardwaj N. Plasmacytoid dendritic cells: linking innate and adaptive immunity. J Virol 2005;79 :17–27.
71. Shirai A, Conover J, Klinman DM. Increased activation and altered ratio of IFNg: IL-4 secreting cells in MRL/lpr mice. Autoimmunity1996; 21 :107-16.
72. Takahashi S, Fossati L, Iwamoto M, Merino R, Motta R, Kobayakawa T, Izui S. Imbalance towards Th1 predominance is associated with acceleration of lupus-like autoimmune syndrome in MRL mice. J Clin Invest 1996; 97 :1597-604.
73. Wang RX, Yu CR, Dambuza IM, Mahdi RM, Dolinska MB, Sergeev YV, Wingfield PT, Kim SH, Egwuagu CE. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med 2014;20 :633-41.
74. Yanaba K, Bouaziz JD, Haas KM, Poe JC, Fujimoto M, Tedder TF. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T Cell-dependent inflammatory responses. Immunity 2008;28 :639-50.
75. Yanaba K, Bouaziz JD, Matsushita T, Tsubata T, Tedder TF. The development and function of regulatory B cells expressing IL-10 (B10 cells) requires antigen receptor diversity and TLR signals. J Immunol 2009; 182 :7459-72.
76. Rosser EC, Blair PA, Mauri C. Cellular targets of regulatory B cell-mediated suppression. Molecular Immunol 2014;62 :296-304.
77. Gray M, Miles K, Salter D, Gray D, Savill J. Apoptotic cells protect mice from autoimmune inflammation by the induction of regulatory B cells. PNAS 2007; 104 :14080-85.
78. Evans JG, Chavez-Rueda KA, Eddaoudi A, Meyer-Bahlburg A, Rawlings DJ, Ehrenstein MR, Mauri C. Novel suppressive function of transitional 2 B cells in experimental arthritis. J Immunol 2007;178 :7868-78.
79. Watanabe R, Ishiura N, Nakashima H, Kuwano Y, Okochi H, Tamaki K, Sato S, Tedder TF, Fujimoto M. Regulatory B cells (B10 cells) have a suppressive role in murine lupus: CD19 and B10 Cell deficiency exacerbates systemic autoimmunity. J Immunol 2010;184 :4801-9.
80. Blair PA, Noreña LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, Mauri C. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic lupus erythematosus patients. Immunity 2010;32 :129-40.
81. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol 2018; 19 :108.
82. Iwata Y, Furuichi K, Kitagawa K, Hara A, Okumura T, Kokubo S, Shimizu K, Sakai N, Sagara A, Kurokawa Y, Ueha S, Matsushima K, Kaneko S, Wada T. Involvement of CD11b+ GR-1low cells in autoimmune disorder in MRL-Fas lpr mouse. Clin Exp Nephrol 2010; 14 :411-7.
83. Fujii W, Ashihara E, Hirai H, Nagahara H, Kajitani N, Fujioka K, Murakami K, Seno T, Yamamoto A, Ishino H, Kohno M, Maekawa T, Kawahito Y. Myeloid-derived suppressor cells play crucial roles in the regulation of mouse collagen-induced arthritis. J Immunol 2013;191 :1073-81.
84. Ioannou M, Alissafi T, Lazaridis I, Deraos G, Matsoukas J, Gravanis A, Mastorodemos V, Plaitakis A, Sharpe A, Boumpas D, Verginis P. Crucial role of granulocytic myeloid-derived suppressor cells in the regulation of central nervous system autoimmune disease. J Immunol 2012;188 :1136.
85. Xia S, Sha H, Yang L, Ji Y, Ostrand-Rosenberg S, Qi L. Gr-1+ CD11b+ Myeloid-derived suppressor cells suppress inflammation and promote insulin sensitivity in obesity. J Biol Chem 2011;286 :23591.
86. Makarenkova VP, Bansal V, Matta BM, Perez LA, Ochoa JB. CD11b+/Gr-1+ Myeloid suppressor cells cause T cell dysfunction after traumatic stress. J Immunol 2006; 176 :2085-94.
87. Park MJ, Lee SH, Kim EK, Lee EJ, Park SH, Kwok SK, Cho ML. Myeloid-derived suppressor cells induce the expansion of regulatory B Cells and ameliorate autoimmunity in the sanroque mouse model of systemic lupus erythematosus. Arthritis Rheumatol 2016;68 :2717-27.
88. Li D, Shi G, Wang J, Zhang D, Pan Y, Dou H, Hou Y. Baicalein ameliorates pristane-induced lupus nephritis via activating Nrf2/HO-1 in myeloid-derived suppressor cells. Arthritis Res Ther 2019;21 :105.
89. Bayik D, Tross D, Klinman DM. Factors influencing the differentiation of human monocytic myeloid-derived suppressor cells into inflammatory macrophages. Front Immunol 2018; 9 :608.
90. Zhan X, Fang Y, Hu S, Wu Y, Yang K, Liao C, Zhang Y, Huang X, Wu M. IFN-gamma differentially regulates subsets of Gr-1(+)CD11b(+) myeloid cells in chronic inflammation. Mol Immunol 2015;66 :451-62.
91. Lian F, Wang Y, Chen J, Xu H, Yang X, Liang L, Zhan Z, Ye Y, Chen M. Activation of farnesoid X receptor attenuates liver injury in systemic lupus erythematosus. Rheumatol Int 2012; 32 :1705-10.
92. Iwata Y, Furuichi K, Sakai N, Yamauchi H, Shinozaki Y, Zhou H, Kurokawa Y, Toyama T, Kitajima S, Okumura T, Yamada S, Maruyama I, Matsushima K, Kaneko S, Wada T. Dendritic cells contribute to autoimmune kidney injury in MRL-Faslpr mice. J Rheumatol 2009;36 :306-14.
93. Yui MA, Brissette WH, Brennan DC, Wuthrich RP, Rubin-Kelley VE. Increased macrophage colony-stimulating factor in neonatal and adult autoimmune MRL-lpr mice. Am J Pathol 1991; 139 :255-61.
94. Bird AK, Chang M, Barnard J, Goldman BI, Meednu N, Rangel-Moreno J, Anolik JH. Neutrophils slow disease progression in murine lupus via modulation of autoreactive germinal centers. J Immunol 2017;199 :458-466.
95. Vlachou K, Mintzas K, Glymenaki M, Ioannou M, Papadaki G, Bertsias GK, Sidiropoulos P, Boumpas DT, Verginis P. Elimination of granulocytic myeloid-derived suppressor cells in lupus-prone mice due to reactive oxygen species-dependent extracellular trap formation. Arthritis Rheumatol 2016; 68 :449-61.