J Rheum Dis.  2018 Apr;25(2):81-99. 10.4078/jrd.2018.25.2.81.

Wolves Trapped in the NETs–The Pathogenesis of Lupus Nephritis

Affiliations
  • 1Division of Internal Medicine, Daejeon Veterans Hospital, Daejeon, Korea.
  • 2Division of Rheumatology, Department of Internal Medicine, Daejeon Rheumatoid and Degenerative Arthritis Center, Chungnam National University Hospital, Chungnam National University College of Medicine, Daejeon, Korea. shimsc@cnuh.co.kr

Abstract

Systemic lupus erythematous (SLE) is a systemic autoimmune disease with multi-organ inflammation caused by the production of pathogenic autoantibodies and immune complexes reflecting a global loss of tolerance. Lupus nephritis (LN) is present in approximately 60% of SLE patients and is considered a major predictor of a poor prognosis. To date, many studies utilizing genomics, transcriptomics, epigenetics, metabolomics, and microbiome have been conducted on a range of animal models and lupus patients to understand the pathogenesis of SLE and LN. Collectively, these studies support the concept that LN is caused by increased cell death, which has not been properly dealt with; abnormal innate immunity; hyperactive adaptive immunity; and genetic variants triggered by a range of environmental factors. This review summarizes the results from studies that contributed strongly to elucidating the pathogenesis of SLE and LN, highlighting the immunological and non-immunological mechanisms.

Keyword

Lupus nephritis; Apoptosis; Autoimmune diseases; Immunology; Lymphocytes

MeSH Terms

Adaptive Immunity
Allergy and Immunology
Antigen-Antibody Complex
Apoptosis
Autoantibodies
Autoimmune Diseases
Cell Death
Epigenomics
Genomics
Humans
Immunity, Innate
Inflammation
Lupus Nephritis*
Lymphocytes
Metabolomics
Microbiota
Models, Animal
Prognosis
Wolves*
Antigen-Antibody Complex
Autoantibodies

Figure

  • Figure 1 Overall pathogenic mechanism of lupus nephritis. HLA: human leukocyte antigen, SLC5A11: solute carrier family 5 member 11, PDGF: platelet-derived growth factor, TNFSF4: tumor necrosis factor ligand superfamily, member 4, ITGAM: integrin alpha M, STAT: signal transducer and activator of transcription, UV: ultraviolet, RNA: ribonucleic acid, DNA: deoxyribonucleic acid, DNase: deoxyribonuclease, pDC: plasmacytoid dendritic cells, TLR: toll-like receptor, STING: stimulator of interferon gene, IFN: interferon, TNF: tumor necrosis factor, IL: interleukin, BAFF: B cell-activating factor, CCL2: chemokine ligand 2, FGF: fibroblast growth factor, Th: helper T cells, Treg: regulatory T cells, TfH: follicular helper T cells, dsDNA: double strand DNA.

  • Figure 2 Potential role of aberrant cell death in the development of systemic lupus erythematous. CRP: C-reactive protein, PTX: pentraxin-related protein, DNase: deoxyribonuclease, NET: neutrophil extracellular trap, DNA: deoxyribonucleic acid, RNA: ribonucleic acid, AMP: adenosine monophosphate, HMGB1: high-mobility group box 1 protein, pDC: plasmacytoid dendritic cells, TLR: toll-like receptor, STING: stimulator of interferon gene, IFN: interferon, Th: helper T cells, Treg: regulatory T cells, TfH: follicular helper T cells.

  • Figure 3 Innate and acquired immunity processes contribute to systemic lupus erythematous. pDC: plasmacytoid dendritic cells, TLR: toll-like receptor, STING: stimulator of interferon gene, IFN: interferon, TNF: tumor necrosis factor, IL: interleukin, BAFF: B cell-activating factor, Treg: regulatory T cells, Th: helper T cells, TfH: follicular helper T cells.

  • Figure 4 Associated genes and environmental factors with the pathogenesis of systemic lupus erythematous. CRP: C-reactive protein, ATG5: α-glucoside transporter 5, TREX1: three prime repair exonuclease 1, UV: ultraviolet, HLA: human leukocyte antigen, FCGR: Fcγ receptor, IRAK: interleukin 1 receptor associated kinase, IRF: IFN regulatory factor, STAT: signal transducer and activator of transcription, ITGAM: integrin α M, TNFAIP: TNF α-induced protein, IFI27: interferon α-inducible protein 27, IFI44L: interferon-induced protein 44-like, LY6E: lymphocyte antigen 6 complex, locus E, IFITM1: interferon induced transmembrane protein 1, BANK1: B cell scaffold protein with ankyrin repeats 1, BLK: B lymphoid tyrosine kinase, ETS1: ETS proto-oncogene 1, PTPN22: protein tyrosine phosphatase, nonreceptor type 22, TNFSF: tumor necrosis factor ligand superfamily, member, IKZF1: IKAROS family zinc finger 1, SLC5A11: solute carrier family 5 member 11, PDGFR: platelet-derived growth factor receptor, NCF1: neutrophil cytosolic factor 1, ACP5: acid phosphatase 5, PRDM1: PR domain zinc finger protein 1.


Reference

1. Shlomchik MJ, Craft JE, Mamula MJ. From T to B and back again: positive feedback in systemic autoimmune disease. Nat Rev Immunol. 2001; 1:147–153.
Article
2. Ortega LM, Schultz DR, Lenz O, Pardo V, Contreras GN. Review: Lupus nephritis: pathologic features, epidemiology and a guide to therapeutic decisions. Lupus. 2010; 19:557–574.
Article
3. Shim JS, Sung YK, Joo YB, Lee HS, Bae SC. Prevalence and incidence of systemic lupus erythematosus in South Korea. Rheumatol Int. 2014; 34:909–917.
Article
4. Hsu CY, Chiu WC, Yang TS, Chen CJ, Chen YC, Lai HM, et al. Age- and gender-related long-term renal outcome in patients with lupus nephritis. Lupus. 2011; 20:1135–1141.
Article
5. Merrell M, Shulman LE. Determination of prognosis in chronic disease, illustrated by systemic lupus erythematosus. J Chronic Dis. 1955; 1:12–32.
Article
6. Pons-Estel GJ, Alarcón GS, Scofield L, Reinlib L, Cooper GS. Understanding the epidemiology and progression of systemic lupus erythematosus. Semin Arthritis Rheum. 2010; 39:257–268.
Article
7. Maroz N, Segal MS. Lupus nephritis and end-stage kidney disease. Am J Med Sci. 2013; 346:319–323.
Article
8. Anaya JM, Cañas C, Mantilla RD, Pineda-Tamayo R, Tobón GJ, Herrera-Diaz C, et al. Lupus nephritis in Colombians: contrasts and comparisons with other populations. Clin Rev Allergy Immunol. 2011; 40:199–207.
Article
9. Yap DY, Tang CS, Ma MK, Lam MF, Chan TM. Survival analysis and causes of mortality in patients with lupus nephritis. Nephrol Dial Transplant. 2012; 27:3248–3254.
Article
10. Lerang K, Gilboe IM, Steinar Thelle D, Gran JT. Mortality and years of potential life loss in systemic lupus erythematosus: a population-based cohort study. Lupus. 2014; 23:1546–1552.
Article
11. Bernatsky S, Boivin JF, Joseph L, Manzi S, Ginzler E, Gladman DD, et al. Mortality in systemic lupus erythematosus. Arthritis Rheum. 2006; 54:2550–2557.
Article
12. Faurschou M, Dreyer L, Kamper AL, Starklint H, Jacobsen S. Long-term mortality and renal outcome in a cohort of 100 patients with lupus nephritis. Arthritis Care Res (Hoboken). 2010; 62:873–880.
Article
13. Pieterse E, van der. Breaking immunological tolerance in systemic lupus erythematosus. Front Immunol. 2014; 5:164.
Article
14. Lynch DH, Watson ML, Alderson MR, Baum PR, Miller RE, Tough T, et al. The mouse Fas-ligand gene is mutated in gld mice and is part of a TNF family gene cluster. Immunity. 1994; 1:131–136.
Article
15. Takahashi T, Tanaka M, Brannan CI, Jenkins NA, Copeland NG, Suda T, et al. Generalized lymphoproliferative disease in mice, caused by a point mutation in the Fas ligand. Cell. 1994; 76:969–976.
Article
16. 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; 356:314–317.
Article
17. Fisher GH, Rosenberg FJ, Straus SE, Dale JK, Middleton LA, Lin AY, et al. Dominant interfering Fas gene mutations impair apoptosis in a human autoimmune lymphoproliferative syndrome. Cell. 1995; 81:935–946.
Article
18. Rieux-Laucat F, Le Deist F, Hivroz C, Roberts IA, Debatin KM, Fischer A, et al. Mutations in Fas associated with human lymphoproliferative syndrome and autoimmunity. Science. 1995; 268:1347–1349.
Article
19. Gaipl US, Voll RE, Sheriff A, Franz S, Kalden JR, Herrmann M. Impaired clearance of dying cells in systemic lupus erythematosus. Autoimmun Rev. 2005; 4:189–194.
Article
20. Muñoz LE, Lauber K, Schiller M, Manfredi AA, Herrmann M. The role of defective clearance of apoptotic cells in systemic autoimmunity. Nat Rev Rheumatol. 2010; 6:280–289.
Article
21. van Bavel CC, Dieker J, Muller S, Briand JP, Monestier M, Berden JH, et al. Apoptosis-associated acetylation on histone H2B is an epitope for lupus autoantibodies. Mol Immunol. 2009; 47:511–516.
Article
22. Dieker JW, Fransen JH, van Bavel CC, Briand JP, Jacobs CW, Muller S, et al. Apoptosis-induced acetylation of histones is pathogenic in systemic lupus erythematosus. Arthritis Rheum. 2007; 56:1921–1933.
Article
23. Plaué S, Muller S, van Regenmortel MH. A branched, synthetic octapeptide of ubiquitinated histone H2A as target of autoantibodies. J Exp Med. 1989; 169:1607–1617.
Article
24. Dieker J, Tel J, Pieterse E, Thielen A, Rother N, Bakker M, et al. Circulating apoptotic microparticles in systemic lupus erythematosus patients drive the activation of dendritic cell subsets and prime neutrophils for NETosis. Arthritis Rheumatol. 2016; 68:462–472.
Article
25. Dieker JW, van der Vlag J, Berden JH. Deranged removal of apoptotic cells: its role in the genesis of lupus. Nephrol Dial Transplant. 2004; 19:282–285.
Article
26. Soto HM, Parra G, Rodríguez-Itrube B. Circulating levels of cytokines in poststreptococcal glomerulonephritis. Clin Nephrol. 1997; 47:6–12.
27. Makino H, Sugiyama H, Yamasaki Y, Maeshima Y, Wada J, Kashihara N. Glomerular cell apoptosis in human lupus nephritis. Virchows Arch. 2003; 443:67–77.
28. Soto H, Mosquera J, Rodríguez-Iturbe B, Henriquez La, Pinto A. Apoptosis in proliferative glomerulonephritis: decreased apoptosis expression in lupus nephritis. Nephrol Dial Transplant. 1997; 12:273–280.
Article
29. Faurschou M, Penkowa M, Andersen CB, Starklint H, Jacobsen S. Renal cell apoptosis in human lupus nephritis: a histological study. Lupus. 2009; 18:994–999.
Article
30. Watanabe M, Hitomi M, van der Wee K, Rothenberg F, Fisher SA, Zucker R, et al. The pros and cons of apoptosis assays for use in the study of cells, tissues, and organs. Microsc Microanal. 2002; 8:375–391.
Article
31. van Bavel CC, Dieker JW, Kroeze Y, Tamboer WP, Voll R, Muller S, et al. Apoptosis-induced histone H3 methylation is targeted by autoantibodies in systemic lupus erythematosus. Ann Rheum Dis. 2011; 70:201–207.
Article
32. Stöckl F, Muller S, Batsford S, Schmiedeke T, Waldherr R, Andrassy K, et al. A role for histones and ubiquitin in lupus nephritis? Clin Nephrol. 1994; 41:10–17.
33. Papayannopoulos V, Metzler KD, Hakkim A, Zychlinsky A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 2010; 191:677–691.
Article
34. Saffarzadeh M, Preissner KT. Fighting against the dark side of neutrophil extracellular traps in disease: manoeuvres for host protection. Curr Opin Hematol. 2013; 20:3–9.
35. Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011; 187:538–552.
Article
36. Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med. 2011; 3:73ra20.
Article
37. Knight JS, Subramanian V, O'Dell AA, Yalavarthi S, Zhao W, Smith CK, et al. Peptidylarginine deiminase inhibition disrupts NET formation and protects against kidney, skin and vascular disease in lupus-prone MRL/lpr mice. Ann Rheum Dis. 2015; 74:2199–2206.
Article
38. Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med. 2011; 3:73ra19.
Article
39. Napirei M, Karsunky H, Zevnik B, Stephan H, Mannherz HG, Möröy T. Features of systemic lupus erythematosus in Dnase1-deficient mice. Nat Genet. 2000; 25:177–181.
Article
40. Skiljevic D, Jeremic I, Nikolic M, Andrejevic S, Sefik-Bukilica M, Stojimirovic B, et al. Serum DNase I activity in systemic lupus erythematosus: correlation with immunoserological markers, the disease activity and organ involvement. Clin Chem Lab Med. 2013; 51:1083–1091.
Article
41. Hakkim A, Fürnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A. 2010; 107:9813–9818.
Article
42. Leffler J, Martin M, Gullstrand B, Tydén H, Lood C, Truedsson L, et al. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol. 2012; 188:3522–3531.
Article
43. Bodaño A, González A, Ferreiros-Vidal I, Balada E, Ordi J, Carreira P, et al. Association of a non-synonymous single-nucleotide polymorphism of DNASEI with SLE susceptibility. Rheumatology (Oxford). 2006; 45:819–823.
44. Theofilopoulos AN, Kono DH, Beutler B, Baccala R. Intracellular nucleic acid sensors and autoimmunity. J Interferon Cytokine Res. 2011; 31:867–886.
Article
45. Celhar T, Magalhães R, Fairhurst AM. TLR7 and TLR9 in SLE: when sensing self goes wrong. Immunol Res. 2012; 53:58–77.
Article
46. Komatsuda A, Wakui H, Iwamoto K, Ozawa M, Togashi M, Masai R, et al. Up-regulated expression of Toll-like receptors mRNAs in peripheral blood mononuclear cells from patients with systemic lupus erythematosus. Clin Exp Immunol. 2008; 152:482–487.
Article
47. Lyn-Cook BD, Xie C, Oates J, Treadwell E, Word B, Hammons G, et al. Increased expression of Toll-like receptors (TLRs) 7 and 9 and other cytokines in systemic lupus erythematosus (SLE) patients: ethnic differences and potential new targets for therapeutic drugs. Mol Immunol. 2014; 61:38–43.
Article
48. Chauhan SK, Singh VV, Rai R, Rai M, Rai G. Distinct autoantibody profiles in systemic lupus erythematosus patients are selectively associated with TLR7 and TLR9 upregulation. J Clin Immunol. 2013; 33:954–964.
Article
49. Papadimitraki ED, Choulaki C, Koutala E, Bertsias G, Tsatsanis C, Gergianaki I, et al. Expansion of toll-like receptor 9-expressing B cells in active systemic lupus erythematosus: implications for the induction and maintenance of the autoimmune process. Arthritis Rheum. 2006; 54:3601–3611.
Article
50. Christensen SR, Kashgarian M, Alexopoulou L, Flavell RA, Akira S, Shlomchik MJ. Toll-like receptor 9 controls anti-DNA autoantibody production in murine lupus. J Exp Med. 2005; 202:321–331.
Article
51. Flür K, Allam R, Zecher D, Kulkarni OP, Lichtnekert J, Schwarz M, et al. Viral RNA induces type I interferon-dependent cytokine release and cell death in mesangial cells via melanoma-differentiation-associated gene-5: Implications for viral infection-associated glomerulonephritis. Am J Pathol. 2009; 175:2014–2022.
52. Allam R, Lichtnekert J, Moll AG, Taubitz A, Vielhauer V, Anders HJ. Viral RNA and DNA trigger common antiviral responses in mesangial cells. J Am Soc Nephrol. 2009; 20:1986–1996.
Article
53. Hägele H, Allam R, Pawar RD, Anders HJ. Double-stranded RNA activates type I interferon secretion in glomerular endothelial cells via retinoic acid-inducible gene (RIG)-1. Nephrol Dial Transplant. 2009; 24:3312–3318.
54. Hägele H, Allam R, Pawar RD, Reichel CA, Krombach F, Anders HJ. Double-stranded DNA activates glomerular endothelial cells and enhances albumin permeability via a toll-like receptor-independent cytosolic DNA recognition pathway. Am J Pathol. 2009; 175:1896–1904.
Article
55. Fairhurst AM, Xie C, Fu Y, Wang A, Boudreaux C, Zhou XJ, et al. Type I interferons produced by resident renal cells may promote end-organ disease in autoantibody-mediated glomerulonephritis. J Immunol. 2009; 183:6831–6838.
Article
56. Anders HJ, Lichtnekert J, Allam R. Interferon-alpha and -beta in kidney inflammation. Kidney Int. 2010; 77:848–854.
57. Triantafyllopoulou A, Franzke CW, Seshan SV, Perino G, Kalliolias GD, Ramanujam M, et al. Proliferative lesions and metalloproteinase activity in murine lupus nephritis mediated by type I interferons and macrophages. Proc Natl Acad Sci U S A. 2010; 107:3012–3017.
Article
58. Anders HJ, Vielhauer V, Eis V, Linde Y, Kretzler M, Perez de Lema G, et al. Activation of toll-like receptor-9 induces progression of renal disease in MRL-Fas(lpr) mice. FASEB J. 2004; 18:534–536.
Article
59. Savarese E, Steinberg C, Pawar RD, Reindl W, Akira S, Anders HJ, et al. Requirement of Toll-like receptor 7 for pristane-induced production of autoantibodies and development of murine lupus nephritis. Arthritis Rheum. 2008; 58:1107–1115.
Article
60. Pawar RD, Patole PS, Zecher D, Segerer S, Kretzler M, Schlöndorff D, et al. Toll-like receptor-7 modulates immune complex glomerulonephritis. J Am Soc Nephrol. 2006; 17:141–149.
Article
61. Pawar RD, Ramanjaneyulu A, Kulkarni OP, Lech M, Segerer S, Anders HJ. Inhibition of Toll-like receptor-7 (TLR-7) or TLR-7 plus TLR-9 attenuates glomerulonephritis and lung injury in experimental lupus. J Am Soc Nephrol. 2007; 18:1721–1731.
Article
62. Subramanian S, Tus K, Li QZ, Wang A, Tian XH, Zhou J, et al. A Tlr7 translocation accelerates systemic autoimmunity in murine lupus. Proc Natl Acad Sci U S A. 2006; 103:9970–9975.
Article
63. Ramirez-Ortiz ZG, Prasad A, Griffith JW, Pendergraft WF 3rd, Cowley GS, Root DE, et al. The receptor TREML4 amplifies TLR7-mediated signaling during antiviral responses and autoimmunity. Nat Immunol. 2015; 16:495–504.
Article
64. Patole PS, Gröne HJ, Segerer S, Ciubar R, Belemezova E, Henger A, et al. Viral double-stranded RNA aggravates lupus nephritis through Toll-like receptor 3 on glomerular mesangial cells and antigen-presenting cells. J Am Soc Nephrol. 2005; 16:1326–1338.
Article
65. Imaizumi T, Aizawa T, Segawa C, Shimada M, Tsuruga K, Kawaguchi S, et al. Toll-like receptor 3 signaling contributes to the expression of a neutrophil chemoattractant, CXCL1 in human mesangial cells. Clin Exp Nephrol. 2015; 19:761–770.
Article
66. Tian J, Ma Y, Li J, Cen H, Wang DG, Feng CC, et al. The TLR7 7926A>G polymorphism is associated with susceptibility to systemic lupus erythematosus. Mol Med Rep. 2012; 6:105–110.
67. Ramachandran R, Sharma V, Rathi M, Yadav AK, Sharma A, Kohli HS, et al. Association between -1486 T>C and +1174 G>A single nucleotide polymorphisms in TLR9 gene and severity of lupus nephritis. Indian J Nephrol. 2012; 22:125–129.
68. Munroe ME, Lu R, Zhao YD, Fife DA, Robertson JM, Guthridge JM, et al. Altered type II interferon precedes autoantibody accrual and elevated type I interferon activity prior to systemic lupus erythematosus classification. Ann Rheum Dis. 2016; 75:2014–2021.
Article
69. Weckerle CE, Mangale D, Franek BS, Kelly JA, Kumabe M, James JA, et al. Large-scale analysis of tumor necrosis factor α levels in systemic lupus erythematosus. Arthritis Rheum. 2012; 64:2947–2952.
70. Yarilina A, Park-Min KH, Antoniv T, Hu X, Ivashkiv LB. TNF activates an IRF1-dependent autocrine loop leading to sustained expression of chemokines and STAT1-dependent type I interferon-response genes. Nat Immunol. 2008; 9:378–387.
Article
71. Flint SM, Jovanovic V, Teo BW, Mak A, Thumboo J, McKinney EF, et al. Leucocyte subset-specific type 1 interferon signatures in SLE and other immune-mediated diseases. RMD Open. 2016; 2:e000183.
Article
72. Liu Z, Bethunaickan R, Huang W, Lodhi U, Solano I, Madaio MP, et al. Interferon-α accelerates murine systemic lupus erythematosus in a T cell-dependent manner. Arthritis Rheum. 2011; 63:219–229.
Article
73. Fairhurst AM, Mathian A, Connolly JE, Wang A, Gray HF, George TA, et al. Systemic IFN-alpha drives kidney nephritis in B6.Sle123 mice. Eur J Immunol. 2008; 38:1948–1960.
74. Solomou EE, Juang YT, Gourley MF, Kammer GM, Tsokos GC. Molecular basis of deficient IL-2 production in T cells from patients with systemic lupus erythematosus. J Immunol. 2001; 166:4216–4222.
Article
75. Li D, Guo B, Wu H, Tan L, Chang C, Lu Q. Interleukin-17 in systemic lupus erythematosus: a comprehensive review. Autoimmunity. 2015; 48:353–361.
Article
76. Kyttaris VC, Kampagianni O, Tsokos GC. Treatment with anti-interleukin 23 antibody ameliorates disease in lupus-prone mice. Biomed Res Int. 2013; 2013:861028.
Article
77. Pers JO, Daridon C, Devauchelle V, Jousse S, Saraux A, Jamin C, et al. BAFF overexpression is associated with autoantibody production in autoimmune diseases. Ann N Y Acad Sci. 2005; 1050:34–39.
Article
78. Stohl W, Xu D, Kim KS, Koss MN, Jorgensen TN, Deocharan B, et al. BAFF overexpression and accelerated glomerular disease in mice with an incomplete genetic predisposition to systemic lupus erythematosus. Arthritis Rheum. 2005; 52:2080–2091.
Article
79. Liu Z, Davidson A. BAFF and selection of autoreactive B cells. Trends Immunol. 2011; 32:388–394.
Article
80. Bethunaickan R, Berthier CC, Zhang W, Eksi R, Li HD, Guan Y, et al. Identification of stage-specific genes associated with lupus nephritis and response to remission induction in (NZB × NZW)F1 and NZM2410 mice. Arthritis Rheumatol. 2014; 66:2246–2258.
Article
81. Pérez de Lema G, Maier H, Nieto E, Vielhauer V, Luckow B, Mampaso F, et al. Chemokine expression precedes inflammatory cell infiltration and chemokine receptor and cytokine expression during the initiation of murine lupus nephritis. J Am Soc Nephrol. 2001; 12:1369–1382.
82. Liu J, Karypis G, Hippen KL, Vegoe AL, Ruiz P, Gilkeson GS, et al. Genomic view of systemic autoimmunity in MRLlpr mice. Genes Immun. 2006; 7:156–168.
Article
83. Berthier CC, Bethunaickan R, Gonzalez-Rivera T, Nair V, Ramanujam M, Zhang W, et al. Cross-species transcriptional network analysis defines shared inflammatory responses in murine and human lupus nephritis. J Immunol. 2012; 189:988–1001.
Article
84. Migliorini A, Angelotti ML, Mulay SR, Kulkarni OO, Demleitner J, Dietrich A, et al. The antiviral cytokines IFN-α and IFN-β modulate parietal epithelial cells and promote podocyte loss: implications for IFN toxicity, viral glomerulonephritis, and glomerular regeneration. Am J Pathol. 2013; 183:431–440.
85. Gurkan S, Cabinian A, Lopez V, Bhaumik M, Chang JM, Rabson AB, et al. Inhibition of type I interferon signalling prevents TLR ligand-mediated proteinuria. J Pathol. 2013; 231:248–256.
Article
86. Tesch GH, Maifert S, Schwarting A, Rollins BJ, Kelley VR. Monocyte chemoattractant protein 1-dependent leukocytic infiltrates are responsible for autoimmune disease in MRL-Fas(lpr) mice. J Exp Med. 1999; 190:1813–1824.
Article
87. Bethunaickan R, Sahu R, Liu Z, Tang YT, Huang W, Edegbe O, et al. Anti-tumor necrosis factor α treatment of interferon-α-induced murine lupus nephritis reduces the renal macrophage response but does not alter glomerular immune complex formation. Arthritis Rheum. 2012; 64:3399–3408.
Article
88. Tucci M, Stucci S, Strippoli S, Silvestris F. Cytokine overproduction, T-cell activation, and defective T-regulatory functions promote nephritis in systemic lupus erythematosus. J Biomed Biotechnol. 2010; 2010:457146.
Article
89. Boswell JM, Yui MA, Burt DW, Kelley VE. Increased tumor necrosis factor and IL-1 beta gene expression in the kidneys of mice with lupus nephritis. J Immunol. 1988; 141:3050–3054.
90. Herrera-Esparza R, Barbosa-Cisneros O, Villalobos-Hurtado R, Avalos-Díaz E. Renal expression of IL-6 and TNFalpha genes in lupus nephritis. Lupus. 1998; 7:154–158.
91. Zhao J, Wang H, Dai C, Wang H, Zhang H, Huang Y, et al. P2X7 blockade attenuates murine lupus nephritis by inhibiting activation of the NLRP3/ASC/caspase 1 pathway. Arthritis Rheum. 2013; 65:3176–3185.
92. Kahlenberg JM, Kaplan MJ. The inflammasome and lupus: another innate immune mechanism contributing to disease pathogenesis? Curr Opin Rheumatol. 2014; 26:475–481.
93. Zhao J, Wang H, Huang Y, Zhang H, Wang S, Gaskin F, et al. Lupus nephritis: glycogen synthase kinase 3β promotion of renal damage through activation of the NLRP3 inflammasome in lupus-prone mice. Arthritis Rheumatol. 2015; 67:1036–1044.
Article
94. Amarilyo G, Lourenço EV, Shi FD, La Cava A. IL-17 promotes murine lupus. J Immunol. 2014; 193:540–543.
Article
95. Schmidt T, Paust HJ, Krebs CF, Turner JE, Kaffke A, Bennstein SB, et al. Function of the Th17/interleukin-17A immune response in murine lupus nephritis. Arthritis Rheumatol. 2015; 67:475–487.
Article
96. Liarski VM, Kaverina N, Chang A, Brandt D, Yanez D, Talasnik L, et al. Cell distance mapping identifies functional T follicular helper cells in inflamed human renal tissue. Sci Transl Med. 2014; 6:230ra46.
Article
97. Lee SK, Silva DG, Martin JL, Pratama A, Hu X, Chang PP, et al. Interferon-γ excess leads to pathogenic accumulation of follicular helper T cells and germinal centers. Immunity. 2012; 37:880–892.
Article
98. Miyake K, Nakashima H, Akahoshi M, Inoue Y, Nagano S, Tanaka Y, et al. Genetically determined interferon-gamma production influences the histological phenotype of lupus nephritis. Rheumatology (Oxford). 2002; 41:518–524.
Article
99. Namjou B, Sestak AL, Armstrong DL, Zidovetzki R, Kelly JA, Jacob N, et al. High-density genotyping of STAT4 reveals multiple haplotypic associations with systemic lupus erythematosus in different racial groups. Arthritis Rheum. 2009; 60:1085–1095.
Article
100. Pickering MC, Botto M, Taylor PR, Lachmann PJ, Walport MJ. Systemic lupus erythematosus, complement deficiency, and apoptosis. Adv Immunol. 2000; 76:227–324.
Article
101. Barilla-LaBarca ML, Atkinson JP. Rheumatic syndromes associated with complement deficiency. Curr Opin Rheumatol. 2003; 15:55–60.
Article
102. Walport MJ, Davies KA, Botto M. C1q and systemic lupus erythematosus. Immunobiology. 1998; 199:265–285.
Article
103. Kallel-Sellami M, Baili-Klila L, Zerzeri Y, Laadhar L, Blouin J, Abdelmalek R, et al. Hereditary complement deficiency and lupus: report of four Tunisian cases. Ann N Y Acad Sci. 2007; 1108:197–202.
Article
104. Mayilyan KR. Complement genetics, deficiencies, and disease associations. Protein Cell. 2012; 3:487–496.
Article
105. Laich A, Sim RB. Cross-talk between the human complement classical and alternative pathways: evidence for a C4bBb 'hybrid' C3 convertase. Mol Immunol. 2001; 38:105.
106. Jönsson G, Sjöholm AG, Truedsson L, Bengtsson AA, Braconier JH, Sturfelt G. Rheumatological manifestations, organ damage and autoimmunity in hereditary C2 deficiency. Rheumatology (Oxford). 2007; 46:1133–1139.
Article
107. Stegert M, Bock M, Trendelenburg M. Clinical presentation of human C1q deficiency: How much of a lupus? Mol Immunol. 2015; 67:3–11.
Article
108. Beurskens FJ, van Schaarenburg RA, Trouw LA. C1q, antibodies and anti-C1q autoantibodies. Mol Immunol. 2015; 68:6–13.
Article
109. Sinico RA, Rimoldi L, Radice A, Bianchi L, Gallelli B, Moroni G. Anti-C1q autoantibodies in lupus nephritis. Ann N Y Acad Sci. 2009; 1173:47–51.
Article
110. Birmingham DJ, Bitter JE, Ndukwe EG, Dials S, Gullo TR, Conroy S, et al. Relationship of circulating anti-C3b and anti-C1q IgG to lupus nephritis and its flare. Clin J Am Soc Nephrol. 2016; 11:47–53.
Article
111. Gargiulo Mde L, Gómez G, Khoury M, Collado MV, Suárez L, Álvarez C, et al. Association between the presence of anti-C1q antibodies and active nephritis in patients with systemic lupus erythematosus. Medicina (B Aires). 2015; 75:23–28.
112. Leffler J, Bengtsson AA, Blom AM. The complement system in systemic lupus erythematosus: an update. Ann Rheum Dis. 2014; 73:1601–1606.
Article
113. Jönsen A, Gunnarsson I, Gullstrand B, Svenungsson E, Bengtsson AA, Nived O, et al. Association between SLE nephritis and polymorphic variants of the CRP and FcgammaRIIIa genes. Rheumatology (Oxford). 2007; 46:1417–1421.
114. Seelen MA, van der Bijl EA, Trouw LA, Zuiverloon TC, Munoz JR, Fallaux-van den Houten FC, et al. A role for mannose-binding lectin dysfunction in generation of autoantibodies in systemic lupus erythematosus. Rheumatology (Oxford). 2005; 44:111–119.
Article
115. Dörner T, Jacobi AM, Lee J, Lipsky PE. Abnormalities of B cell subsets in patients with systemic lupus erythematosus. J Immunol Methods. 2011; 363:187–197.
Article
116. Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science. 2003; 301:1374–1377.
Article
117. Yurasov S, Tiller T, Tsuiji M, Velinzon K, Pascual V, Wardemann H, et al. Persistent expression of autoantibodies in SLE patients in remission. J Exp Med. 2006; 203:2255–2261.
Article
118. Guerrier T, Youinou P, Pers JO, Jamin C. TLR9 drives the development of transitional B cells towards the marginal zone pathway and promotes autoimmunity. J Autoimmun. 2012; 39:173–179.
Article
119. Thien M, Phan TG, Gardam S, Amesbury M, Basten A, Mackay F, et al. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity. 2004; 20:785–798.
Article
120. Lesley R, Xu Y, Kalled SL, Hess DM, Schwab SR, Shu HB, et al. Reduced competitiveness of autoantigen-engaged B cells due to increased dependence on BAFF. Immunity. 2004; 20:441–453.
Article
121. Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M, Schneider P, et al. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med. 1999; 190:1697–1710.
Article
122. Gao N, Dresel J, Eckstein V, Gellert R, Störch H, Venigalla RK, et al. Impaired suppressive capacity of activation-induced regulatory B cells in systemic lupus erythematosus. Arthritis Rheumatol. 2014; 66:2849–2861.
Article
123. Pillai S, Mattoo H, Cariappa A. B cells and autoimmunity. Curr Opin Immunol. 2011; 23:721–731.
Article
124. Bohnhorst JØ, Bjørgan MB, Thoen JE, Natvig JB, Thompson KM. Bm1-Bm5 classification of peripheral blood B cells reveals circulating germinal center founder cells in healthy individuals and disturbance in the B cell subpopulations in patients with primary Sjögren's syndrome. J Immunol. 2001; 167:3610–3618.
Article
125. Harada Y, Kawano MM, Huang N, Mahmoud MS, Lisukov IA, Mihara K, et al. Identification of early plasma cells in peripheral blood and their clinical significance. Br J Haematol. 1996; 92:184–191.
Article
126. Nimmerjahn F, Ravetch JV. Fc-receptors as regulators of immunity. Adv Immunol. 2007; 96:179–204.
Article
127. Ettinger R, Kuchen S, Lipsky PE. Interleukin 21 as a target of intervention in autoimmune disease. Ann Rheum Dis. 2008; 67:Suppl 3. iii83–iii86.
Article
128. Goilav B, Putterman C. The role of Anti-DNA antibodies in the development of lupus nephritis: a complementary, or alternative, viewpoint? Semin Nephrol. 2015; 35:439–443.
Article
129. Sabbaga J, Pankewycz OG, Lufft V, Schwartz RS, Madaio MP. Cross-reactivity distinguishes serum and nephritogenic anti-DNA antibodies in human lupus from their natural counterparts in normal serum. J Autoimmun. 1990; 3:215–235.
Article
130. Xie C, Liang Z, Chang S, Mohan C. Use of a novel elution regimen reveals the dominance of polyreactive antinuclear autoantibodies in lupus kidneys. Arthritis Rheum. 2003; 48:2343–2352.
Article
131. Kalaaji M, Sturfelt G, Mjelle JE, Nossent H, Rekvig OP. Critical comparative analyses of anti-alpha-actinin and glomerulus-bound antibodies in human and murine lupus nephritis. Arthritis Rheum. 2006; 54:914–926.
132. Mannik M, Merrill CE, Stamps LD, Wener MH. Multiple autoantibodies form the glomerular immune deposits in patients with systemic lupus erythematosus. J Rheumatol. 2003; 30:1495–1504.
133. Seredkina N, Van Der Vlag J, Berden J, Mortensen E, Rekvig OP. Lupus nephritis: enigmas, conflicting models and an emerging concept. Mol Med. 2013; 19:161–169.
Article
134. Fenton K, Fismen S, Hedberg A, Seredkina N, Fenton C, Mortensen ES, et al. Anti-dsDNA antibodies promote initiation, and acquired loss of renal Dnase1 promotes progression of lupus nephritis in autoimmune (NZBxNZW)F1 mice. PLoS One. 2009; 4:e8474.
Article
135. Seredkina N, Rekvig OP. Acquired loss of renal nuclease activity is restricted to DNaseI and is an organ-selective feature in murine lupus nephritis. Am J Pathol. 2011; 179:1120–1128.
Article
136. Rekvig OP. The anti-DNA antibody: origin and impact, dogmas and controversies. Nat Rev Rheumatol. 2015; 11:530–540.
Article
137. Adu D, Dobson J, Williams DG. DNA-anti-DNA circulating complexes in the nephritis of systemic lupus erythematosus. Clin Exp Immunol. 1981; 43:605–614.
138. Fenton KA, Tømmerås B, Marion TN, Rekvig OP. Pure anti-dsDNA mAbs need chromatin structures to promote glomerular mesangial deposits in BALB/c mice. Autoimmunity. 2010; 43:179–188.
Article
139. Ehrenstein MR, Katz DR, Griffiths MH, Papadaki L, Winkler TH, Kalden JR, et al. Human IgG anti-DNA antibodies deposit in kidneys and induce proteinuria in SCID mice. Kidney Int. 1995; 48:705–711.
Article
140. Bruschi M, Sinico RA, Moroni G, Pratesi F, Migliorini P, Galetti M, et al. Glomerular autoimmune multicomponents of human lupus nephritis in vivo: α-enolase and annexin AI. J Am Soc Nephrol. 2014; 25:2483–2498.
Article
141. Chang A, Henderson SG, Brandt D, Liu N, Guttikonda R, Hsieh C, et al. In situ B cell-mediated immune responses and tubulointerstitial inflammation in human lupus nephritis. J Immunol. 2011; 186:1849–1860.
Article
142. Espeli M, Bökers S, Giannico G, Dickinson HA, Bardsley V, Fogo AB, et al. Local renal autoantibody production in lupus nephritis. J Am Soc Nephrol. 2011; 22:296–305.
Article
143. Kinloch AJ, Chang A, Ko K, Henry Dunand CJ, Henderson S, et al. Vimentin is a dominant target of in situ humoral immunity in human lupus tubulointerstitial nephritis. Arthritis Rheumatol. 2014; 66:3359–3370.
Article
144. Rose ML. Role of anti-vimentin antibodies in allograft rejection. Hum Immunol. 2013; 74:1459–1462.
Article
145. Packard TA, Cambier JC. B lymphocyte antigen receptor signaling: initiation, amplification, and regulation. F1000Prime Rep. 2013; 5:40.
Article
146. Manjarrez-Orduño N, Marasco E, Chung SA, Katz MS, Kiridly JF, Simpfendorfer KR, et al. CSK regulatory polymorphism is associated with systemic lupus erythematosus and influences B-cell signaling and activation. Nat Genet. 2012; 44:1227–1230.
Article
147. Lu R, Vidal GS, Kelly JA, Delgado-Vega AM, Howard XK, Macwana SR, et al. Genetic associations of LYN with systemic lupus erythematosus. Genes Immun. 2009; 10:397–403.
Article
148. Patole PS, Zecher D, Pawar RD, Gröne HJ, Schlöndorff D, Anders HJ. G-rich DNA suppresses systemic lupus. J Am Soc Nephrol. 2005; 16:3273–3280.
Article
149. Peters AL, Plenge RM, Graham RR, Altshuler DM, Moser KL, Gaffney PM, et al. A novel polymorphism of the human CD40 receptor with enhanced function. Blood. 2008; 112:1863–1871.
Article
150. Enyedy EJ, Nambiar MP, Liossis SN, Dennis G, Kammer GM, Tsokos GC. Fc epsilon receptor type I gamma chain replaces the deficient T cell receptor zeta chain in T cells of patients with systemic lupus erythematosus. Arthritis Rheum. 2001; 44:1114–1121.
151. Liossis SN, Ding XZ, Dennis GJ, Tsokos GC. Altered pattern of TCR/CD3-mediated protein-tyrosyl phosphorylation in T cells from patients with systemic lupus erythematosus. Deficient expression of the T cell receptor zeta chain. J Clin Invest. 1998; 101:1448–1457.
Article
152. Kyttaris VC, Juang YT, Tenbrock K, Weinstein A, Tsokos GC. Cyclic adenosine 5′-monophosphate response element modulator is responsible for the decreased expression of c-fos and activator protein-1 binding in T cells from patients with systemic lupus erythematosus. J Immunol. 2004; 173:3557–3563.
Article
153. Crispín JC, Oukka M, Bayliss G, Cohen RA, Van Beek CA, Stillman IE, et al. Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol. 2008; 181:8761–8766.
Article
154. Crispín JC, Tsokos GC. Human TCR-alpha beta+ CD4−CD8− T cells can derive from CD8+ T cells and display an inflammatory effector phenotype. J Immunol. 2009; 183:4675–4681.
155. Shivakumar S, Tsokos GC, Datta SK. T cell receptor alpha/beta expressing double-negative (CD4−/CD8−) and CD4+ T helper cells in humans augment the production of pathogenic anti-DNA autoantibodies associated with lupus nephritis. J Immunol. 1989; 143:103–112.
156. Apostolidis SA, Crispín JC, Tsokos GC. IL-17-producing T cells in lupus nephritis. Lupus. 2011; 20:120–124.
Article
157. Winchester R, Wiesendanger M, Zhang HZ, Steshenko V, Peterson K, Geraldino-Pardilla L, et al. Immunologic characteristics of intrarenal T cells: trafficking of expanded CD8+ T cell β-chain clonotypes in progressive lupus nephritis. Arthritis Rheum. 2012; 64:1589–1600.
Article
158. Enghard P, Rieder C, Kopetschke K, Klocke JR, Undeutsch R, Biesen R, et al. Urinary CD4 T cells identify SLE patients with proliferative lupus nephritis and can be used to monitor treatment response. Ann Rheum Dis. 2014; 73:277–283.
Article
159. Mandik-Nayak L, Seo SJ, Sokol C, Potts KM, Bui A, Erikson J. MRL-lpr/lpr mice exhibit a defect in maintaining developmental arrest and follicular exclusion of anti-double-stranded DNA B cells. J Exp Med. 1999; 189:1799–1814.
Article
160. Sinai P, Dozmorov IM, Song R, Schwartzberg PL, Wakeland EK, Wülfing C. T/B-cell interactions are more transient in response to weak stimuli in SLE-prone mice. Eur J Immunol. 2014; 44:3522–3531.
Article
161. Choi JY, Ho JH, Pasoto SG, Bunin V, Kim ST, Carrasco S, et al. Circulating follicular helper-like T cells in systemic lupus erythematosus: association with disease activity. Arthritis Rheumatol. 2015; 67:988–999.
Article
162. Szabó K, Papp G, Szántó A, Tarr T, Zeher M. A comprehensive investigation on the distribution of circulating follicular T helper cells and B cell subsets in primary Sjögren's syndrome and systemic lupus erythematosus. Clin Exp Immunol. 2016; 183:76–89.
Article
163. Le Coz C, Joublin A, Pasquali JL, Korganow AS, Dumortier H, Monneaux F. Circulating TFH subset distribution is strongly affected in lupus patients with an active disease. PLoS One. 2013; 8:e75319.
Article
164. Jacquemin C, Schmitt N, Contin-Bordes C, Liu Y, Narayanan P, Seneschal J, et al. OX40 Ligand Contributes to Human Lupus Pathogenesis by Promoting T Follicular Helper Response. Immunity. 2015; 42:1159–1170.
Article
165. Cortini A, Ellinghaus U, Malik TH, Cunninghame Graham DS, Botto M, Vyse TJ. B cell OX40L supports T follicular helper cell development and contributes to SLE pathogenesis. Ann Rheum Dis. 2017; 76:2095–2103.
Article
166. Simpson N, Gatenby PA, Wilson A, Malik S, Fulcher DA, Tangye SG, et al. Expansion of circulating T cells resembling follicular helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus. Arthritis Rheum. 2010; 62:234–244.
Article
167. Floege J, Eitner F, Alpers CE. A new look at platelet-derived growth factor in renal disease. J Am Soc Nephrol. 2008; 19:12–23.
Article
168. Fu J, Lee K, Chuang PY, Liu Z, He JC. Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol. 2015; 308:F287–F297.
Article
169. Daehn I, Casalena G, Zhang T, Shi S, Fenninger F, Barasch N, et al. Endothelial mitochondrial oxidative stress determines podocyte depletion in segmental glomerulosclerosis. J Clin Invest. 2014; 124:1608–1621.
Article
170. Schrimpf C, Teebken OE, Wilhelmi M, Duffield JS. The role of pericyte detachment in vascular rarefaction. J Vasc Res. 2014; 51:247–258.
Article
171. Dimke H, Sparks MA, Thomson BR, Frische S, Coffman TM, Quaggin SE. Tubulovascular cross-talk by vascular endothelial growth factor a maintains peritubular microvasculature in kidney. J Am Soc Nephrol. 2015; 26:1027–1038.
Article
172. Kida Y, Ieronimakis N, Schrimpf C, Reyes M, Duffield JS. EphrinB2 reverse signaling protects against capillary rarefaction and fibrosis after kidney injury. J Am Soc Nephrol. 2013; 24:559–572.
Article
173. Kümpers P, David S, Haubitz M, Hellpap J, Horn R, Bröcker V, et al. The Tie2 receptor antagonist angiopoietin 2 facilitates vascular inflammation in systemic lupus erythematosus. Ann Rheum Dis. 2009; 68:1638–1643.
174. Kida Y, Tchao BN, Yamaguchi I. Peritubular capillary rarefaction: a new therapeutic target in chronic kidney disease. Pediatr Nephrol. 2014; 29:333–342.
Article
175. Gilkeson GS, Mashmoushi AK, Ruiz P, Caza TN, Perl A, Oates JC. Endothelial nitric oxide synthase reduces crescentic and necrotic glomerular lesions, reactive oxygen production, and MCP1 production in murine lupus nephritis. PLoS One. 2013; 8:e64650.
Article
176. Van Linthout S, Miteva K, Tschöpe C. Crosstalk between fibroblasts and inflammatory cells. Cardiovasc Res. 2014; 102:258–269.
Article
177. Duffield JS. Cellular and molecular mechanisms in kidney fibrosis. J Clin Invest. 2014; 124:2299–2306.
Article
178. Deapen D, Escalante A, Weinrib L, Horwitz D, Bachman B, Roy-Burman P, et al. A revised estimate of twin concordance in systemic lupus erythematosus. Arthritis Rheum. 1992; 35:311–318.
179. Nishino H, Shibuya K, Nishida Y, Mushimoto M. Lupus erythematosus-like syndrome with selective complete deficiency of C1q. Ann Intern Med. 1981; 95:322–324.
Article
180. Hannema AJ, Kluin-Nelemans JC, Hack CE, Eerenberg-Belmer AJ, Mallée C, van Helden HP. SLE like syndrome and functional deficiency of C1q in members of a large family. Clin Exp Immunol. 1984; 55:106–114.
181. Botto M, Dell'Agnola C, Bygrave AE, Thompson EM, Cook HT, Petry F, et al. Homozygous C1q deficiency causes glomerulonephritis associated with multiple apoptotic bodies. Nat Genet. 1998; 19:56–59.
Article
182. Botto M, Kirschfink M, Macor P, Pickering MC, Würzner R, Tedesco F. Complement in human diseases: Lessons from complement deficiencies. Mol Immunol. 2009; 46:2774–2783.
Article
183. Niu Z, Zhang P, Tong Y. Value of HLA-DR genotype in systemic lupus erythematosus and lupus nephritis: a meta-analysis. Int J Rheum Dis. 2015; 18:17–28.
Article
184. Kim K, Bang SY, Yoo DH, Cho SK, Choi CB, Sung YK, et al. Imputing Variants in HLA-DR beta genes reveals that HLA-DRB1 is solely associated with rheumatoid arthritis and systemic lupus erythematosus. PLoS One. 2016; 11:e0150283.
Article
185. Kim-Howard X, Maiti AK, Anaya JM, Bruner GR, Brown E, Merrill JT, et al. ITGAM coding variant (rs1143679) influences the risk of renal disease, discoid rash and immunological manifestations in patients with systemic lupus erythematosus with European ancestry. Ann Rheum Dis. 2010; 69:1329–1332.
Article
186. Dong C, Ptacek TS, Redden DT, Zhang K, Brown EE, Edberg JC, et al. Fcγ receptor IIIa single-nucleotide polymorphisms and haplotypes affect human IgG binding and are associated with lupus nephritis in African Americans. Arthritis Rheumatol. 2014; 66:1291–1299.
Article
187. Faridi MH, Khan SQ, Zhao W, Lee HW, Altintas MM, Zhang K, et al. CD11b activation suppresses TLR-dependent inflammation and autoimmunity in systemic lupus erythematosus. J Clin Invest. 2017; 127:1271–1283.
Article
188. International MHC, Rioux JD, Goyette P, Vyse TJ, Hammarström L, Fernando MM, et al. Mapping of multiple susceptibility variants within the MHC region for 7 immune-mediated diseases. Proc Natl Acad Sci U S A. 2009; 106:18680–18685.
189. Bentham J, Morris DL, Graham DSC, Pinder CL, Tombleson P, Behrens TW, et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat Genet. 2015; 47:1457–1464.
Article
190. Niewold TB, Kelly JA, Flesch MH, Espinoza LR, Harley JB, Crow MK. Association of the IRF5 risk haplotype with high serum interferon-alpha activity in systemic lupus erythematosus patients. Arthritis Rheum. 2008; 58:2481–2487.
191. Kariuki SN, Franek BS, Kumar AA, Arrington J, Mikolaitis RA, Utset TO, et al. Trait-stratified genome-wide association study identifies novel and diverse genetic associations with serologic and cytokine phenotypes in systemic lupus erythematosus. Arthritis Res Ther. 2010; 12:R151.
Article
192. Ramos PS, Williams AH, Ziegler JT, Comeau ME, Guy RT, Lessard CJ, et al. Genetic analyses of interferon pathway-related genes reveal multiple new loci associated with systemic lupus erythematosus. Arthritis Rheum. 2011; 63:2049–2057.
Article
193. Boone DL, Turer EE, Lee EG, Ahmad RC, Wheeler MT, Tsui C, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004; 5:1052–1060.
Article
194. de Jong TD, Vosslamber S, Mantel E, de Ridder S, Wesseling JG, van der Pouw Kraan TC, et al. Physiological evidence for diversification of IFNα- and IFNβ-mediated response programs in different autoimmune diseases. Arthritis Res Ther. 2016; 18:49.
Article
195. Deng Y, Tsao BP. Advances in lupus genetics and epigenetics. Curr Opin Rheumatol. 2014; 26:482–492.
Article
196. Lessard CJ, Sajuthi S, Zhao J, Kim K, Ice JA, Li H, et al. Identification of a systemic lupus erythematosus risk locus spanning ATG16L2, FCHSD2, and P2RY2 in Koreans. Arthritis Rheumatol. 2016; 68:1197–1209.
197. Sun C, Molineros JE, Looger LL, Zhou XJ, Kim K, Okada Y, et al. High-density genotyping of immune-related loci identifies new SLE risk variants in individuals with Asian ancestry. Nat Genet. 2016; 48:323–330.
Article
198. Olsson LM, Johansson ÅC, Gullstrand B, Jönsen A, Saevarsdottir S, Rönnblom L, et al. A single nucleotide polymorphism in the NCF1 gene leading to reduced oxidative burst is associated with systemic lupus erythematosus. Ann Rheum Dis. 2017; 76:1607–1613.
199. An J, Briggs TA, Dumax-Vorzet A, Alarcón-Riquelme ME, Belot A, Beresford M, et al. Tartrate-Resistant Acid Phosphatase Deficiency in the Predisposition to Systemic Lupus Erythematosus. Arthritis Rheumatol. 2017; 69:131–142.
Article
200. Kim SJ, Schätzle S, Ahmed SS, Haap W, Jang SH, Gregersen PK, et al. Increased cathepsin S in Prdm1-/- dendritic cells alters the TFH cell repertoire and contributes to lupus. Nat Immunol. 2017; 18:1016–1024.
Article
201. International Consortium for Systemic Lupus Erythematosus Genetics (SLEGEN). Harley JB, Alarcón-Riquelme ME, Criswell LA, Jacob CO, Kimberly RP, et al. Genome-wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet. 2008; 40:204–210.
Article
202. Raffler J, Friedrich N, Arnold M, Kacprowski T, Rueedi R, Altmaier E, et al. Genome-wide association study with targeted and non-targeted NMR metabolomics identifies 15 novel loci of urinary human metabolic individuality. PLoS Genet. 2015; 11:e1005487.
Article
203. Tsai LJ, Hsiao SH, Tsai LM, Lin CY, Tsai JJ, Liou DM, et al. The sodium-dependent glucose cotransporter SLC5A11 as an autoimmune modifier gene in SLE. Tissue Antigens. 2008; 71:114–126.
Article
204. Caster DJ, Korte EA, Nanda SK, McLeish KR, Oliver RK, G'sell RT, et al. ABIN1 dysfunction as a genetic basis for lupus nephritis. J Am Soc Nephrol. 2013; 24:1743–1754.
205. Sanchez E, Nadig A, Richardson BC, Freedman BI, Kaufman KM, Kelly JA, et al. Phenotypic associations of genetic susceptibility loci in systemic lupus erythematosus. Ann Rheum Dis. 2011; 70:1752–1757.
Article
206. Bolin K, Sandling JK, Zickert A, Jönsen A, Sjöwall C, Svenungsson E, et al. Association of STAT4 polymorphism with severe renal insufficiency in lupus nephritis. PLoS One. 2013; 8:e84450.
Article
207. Liu K, Li QZ, Delgado-Vega AM, Abelson AK, Sánchez E, Kelly JA, et al. Kallikrein genes are associated with lupus and glomerular basement membrane-specific antibody-induced nephritis in mice and humans. J Clin Invest. 2009; 119:911–923.
208. Lee YH, Bae SC. Association between the functional ITGAM rs1143679 G/A polymorphism and systemic lupus erythematosus/lupus nephritis or rheumatoid arthritis: an update meta-analysis. Rheumatol Int. 2015; 35:815–823.
Article
209. Sawalha AH, Jeffries M, Webb R, Lu Q, Gorelik G, Ray D, et al. Defective T-cell ERK signaling induces interferon-regulated gene expression and overexpression of methylation-sensitive genes similar to lupus patients. Genes Immun. 2008; 9:368–378.
Article
210. Coit P, Renauer P, Jeffries MA, Merrill JT, McCune WJ, Maksimowicz-McKinnon K, et al. Renal involvement in lupus is characterized by unique DNA methylation changes in naïve CD4+ T cells. J Autoimmun. 2015; 61:29–35.
Article
211. Coit P, Dozmorov MG, Merrill JT, McCune WJ, Maksimowicz-McKinnon K, Wren JD, et al. Epigenetic Reprogramming in Naive CD4+ T Cells Favoring T Cell Activation and Non-Th1 Effector T Cell Immune Response as an Early Event in Lupus Flares. Arthritis Rheumatol. 2016; 68:2200–2209.
Article
212. Yang Y, Tang Q, Zhao M, Liang G, Wu H, Li D, et al. The effect of mycophenolic acid on epigenetic modifications in lupus CD4+T cells. Clin Immunol. 2015; 158:67–76.
Article
213. Zhang Z, Song L, Maurer K, Petri MA, Sullivan KE. Global H4 acetylation analysis by ChIP-chip in systemic lupus erythematosus monocytes. Genes Immun. 2010; 11:124–133.
Article
214. Dai Y, Huang YS, Tang M, Lv TY, Hu CX, Tan YH, et al. Microarray analysis of microRNA expression in peripheral blood cells of systemic lupus erythematosus patients. Lupus. 2007; 16:939–946.
Article
215. Dai Y, Sui W, Lan H, Yan Q, Huang H, Huang Y. Comprehensive analysis of microRNA expression patterns in renal biopsies of lupus nephritis patients. Rheumatol Int. 2009; 29:749–754.
Article
216. Costa-Reis P, Russo PA, Zhang Z, Colonna L, Maurer K, Gallucci S, et al. The Role of MicroRNAs and human epidermal growth factor receptor 2 in proliferative lupus nephritis. Arthritis Rheumatol. 2015; 67:2415–2426.
Article
217. Carlsen AL, Schetter AJ, Nielsen CT, Lood C, Knudsen S, Voss A, et al. Circulating microRNA expression profiles associated with systemic lupus erythematosus. Arthritis Rheum. 2013; 65:1324–1334.
Article
218. Yan S, Yim LY, Lu L, Lau CS, Chan VS. MicroRNA regulation in systemic lupus erythematosus pathogenesis. Immune Netw. 2014; 14:138–148.
Article
219. Nelson P, Rylance P, Roden D, Trela M, Tugnet N. Viruses as potential pathogenic agents in systemic lupus erythematosus. Lupus. 2014; 23:596–605.
Article
220. Sawalha AH, Schmid WR, Binder SR, Bacino DK, Harley JB. Association between systemic lupus erythematosus and Helicobacter pylori seronegativity. J Rheumatol. 2004; 31:1546–1550.
221. Ram M, Anaya JM, Barzilai O, Izhaky D, Porat Katz BS, Blank M, et al. The putative protective role of hepatitis B virus (HBV) infection from autoimmune disorders. Autoimmun Rev. 2008; 7:621–625.
Article
222. Chen M, Aosai F, Norose K, Mun HS, Ishikura H, Hirose S, et al. Toxoplasma gondii infection inhibits the development of lupus-like syndrome in autoimmune (New Zealand Black x New Zealand White) F1 mice. Int Immunol. 2004; 16:937–946.
Article
223. Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol. 2005; 23:307–336.
224. Zandman-Goddard G, Berkun Y, Barzilai O, Boaz M, Blank M, Ram M, et al. Exposure to Epstein-Barr virus infection is associated with mild systemic lupus erythematosus disease. Ann N Y Acad Sci. 2009; 1173:658–663.
Article
225. Shi L, Zhang Z, Yu AM, Wang W, Wei Z, Akhter E, et al. The SLE transcriptome exhibits evidence of chronic endotoxin exposure and has widespread dysregulation of noncoding and coding RNAs. PLoS One. 2014; 9:e93846.
Article
226. Nockher WA, Wigand R, Schoeppe W, Scherberich JE. Elevated levels of soluble CD14 in serum of patients with systemic lupus erythematosus. Clin Exp Immunol. 1994; 96:15–19.
227. Gallo PM, Rapsinski GJ, Wilson RP, Oppong GO, Sriram U, Goulian M, et al. Amyloid-DNA Composites of Bacterial Biofilms Stimulate Autoimmunity. Immunity. 2015; 42:1171–1184.
Article
228. Wagner H. Bacterial CpG DNA activates immune cells to signal infectious danger. Adv Immunol. 1999; 73:329–368.
Article
229. Lövgren T, Eloranta ML, Båve U, Alm GV, Rönnblom L. Induction of interferon-alpha production in plasmacytoid dendritic cells by immune complexes containing nucleic acid released by necrotic or late apoptotic cells and lupus IgG. Arthritis Rheum. 2004; 50:1861–1872.
230. Means TK, Latz E, Hayashi F, Murali MR, Golenbock DT, Luster AD. Human lupus autoantibody-DNA complexes activate DCs through cooperation of CD32 and TLR9. J Clin Invest. 2005; 115:407–417.
Article
231. Hevia A, Milani C, López P, Cuervo A, Arboleya S, Duranti S, et al. Intestinal dysbiosis associated with systemic lupus erythematosus. MBio. 2014; 5:e01548–e01514.
Article
232. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013; 504:446–450.
Article
233. Arpaia N, Campbell C, Fan X, Dikiy S, van der, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013; 504:451–455.
Article
234. Pawar RD, Castrezana-Lopez L, Allam R, Kulkarni OP, Segerer S, Radomska E, et al. Bacterial lipopeptide triggers massive albuminuria in murine lupus nephritis by activating Toll-like receptor 2 at the glomerular filtration barrier. Immunology. 2009; 128:1 Suppl. e206–e221.
Article
235. Achtman JC, Werth VP. Pathophysiology of cutaneous lupus erythematosus. Arthritis Res Ther. 2015; 17:182.
Article
236. Caricchio R, McPhie L, Cohen PL. Ultraviolet B radiation-induced cell death: critical role of ultraviolet dose in inflammation and lupus autoantigen redistribution. J Immunol. 2003; 171:5778–5786.
Article
237. Wu Z, Li X, Qin H, Zhu X, Xu J, Shi W. Ultraviolet B enhances DNA hypomethylation of CD4+ T cells in systemic lupus erythematosus via inhibiting DNMT1 catalytic activity. J Dermatol Sci. 2013; 71:167–173.
Article
238. Zhu X, Liang J, Li F, Yang Y, Xiang L, Xu J. Analysis of associations between the patterns of global DNA hypomethylation and expression of DNA methyltransferase in patients with systemic lupus erythematosus. Int J Dermatol. 2011; 50:697–704.
Article
239. Reefman E, Kuiper H, Limburg PC, Kallenberg CG, Bijl M. Type I interferons are involved in the development of ultraviolet B-induced inflammatory skin lesions in systemic lupus erythaematosus patients. Ann Rheum Dis. 2008; 67:11–18.
Article
240. Cornacchia E, Golbus J, Maybaum J, Strahler J, Hanash S, Richardson B. Hydralazine and procainamide inhibit T cell DNA methylation and induce autoreactivity. J Immunol. 1988; 140:2197–2200.
241. Richardson B, Scheinbart L, Strahler J, Gross L, Hanash S, Johnson M. Evidence for impaired T cell DNA methylation in systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum. 1990; 33:1665–1673.
Article
242. Dorgham K, Amoura Z, Parizot C, Arnaud L, Frances C, Pionneau C, et al. Ultraviolet light converts propranolol, a nonselective β-blocker and potential lupus-inducing drug, into a proinflammatory AhR ligand. Eur J Immunol. 2015; 45:3174–3187.
Article
243. Finckh A, Cooper GS, Chibnik LB, Costenbader KH, Watts J, Pankey H, et al. Occupational silica and solvent exposures and risk of systemic lupus erythematosus in urban women. Arthritis Rheum. 2006; 54:3648–3654.
Article
244. Barbhaiya M, Tedeschi SK, Lu B, Malspeis S, Kreps D, Sparks JA, et al. Cigarette smoking and the risk of systemic lupus erythematosus, overall and by anti-double stranded DNA antibody subtype, in the Nurses' Health Study cohorts. Ann Rheum Dis. 2018; 77:196–202.
Article
245. Young KA, Munroe ME, Guthridge JM, Kamen DL, Niewold TB, Gilkeson GS, et al. Combined role of vitamin D status and CYP24A1 in the transition to systemic lupus erythematosus. Ann Rheum Dis. 2017; 76:153–158.
Article
246. Barbhaiya M, Lu B, Sparks JA, Malspeis S, Chang SC, Karlson EW, et al. Influence of alcohol consumption on the risk of systemic lupus erythematosus among women in the nurses' health study cohorts. Arthritis Care Res (Hoboken). 2017; 69:384–392.
Article
247. Dayan M, Zinger H, Kalush F, Mor G, Amir-Zaltzman Y, Kohen F, et al. The beneficial effects of treatment with tamoxifen and anti-oestradiol antibody on experimental systemic lupus erythematosus are associated with cytokine modulations. Immunology. 1997; 90:101–108.
Article
248. Roubinian JR, Talal N, Greenspan JS, Goodman JR, Siiteri PK. Effect of castration and sex hormone treatment on survival, anti-nucleic acid antibodies, and glomerulonephritis in NZB/NZW F1 mice. J Exp Med. 1978; 147:1568–1583.
Article
249. Roubinian J, Talal N, Siiteri PK, Sadakian JA. Sex hormone modulation of autoimmunity in NZB/NZW mice. Arthritis Rheum. 1979; 22:1162–1169.
Article
250. Bynoe MS, Grimaldi CM, Diamond B. Estrogen up-regulates Bcl-2 and blocks tolerance induction of naive B cells. Proc Natl Acad Sci U S A. 2000; 97:2703–2708.
Article
251. Grimaldi CM, Michael DJ, Diamond B. Cutting edge: expansion and activation of a population of autoreactive marginal zone B cells in a model of estrogen-induced lupus. J Immunol. 2001; 167:1886–1890.
Article
252. Grimaldi CM. Sex and systemic lupus erythematosus: the role of the sex hormones estrogen and prolactin on the regulation of autoreactive B cells. Curr Opin Rheumatol. 2006; 18:456–461.
Article
253. Grimaldi CM, Cleary J, Dagtas AS, Moussai D, Diamond B. Estrogen alters thresholds for B cell apoptosis and activation. J Clin Invest. 2002; 109:1625–1633.
Article
254. Hartley SB, Cooke MP, Fulcher DA, Harris AW, Cory S, Basten A, et al. Elimination of self-reactive B lymphocytes proceeds in two stages: arrested development and cell death. Cell. 1993; 72:325–335.
Article
255. Treadwell EL, Wiley K, Word B, Melchior W, Tolleson WH, Gopee N, et al. Prolactin and Dehydroepiandrosterone Levels in Women with Systemic Lupus Erythematosus: The Role of the Extrapituitary Prolactin Promoter Polymorphism at -1149G/T. J Immunol Res. 2015; 2015:435658.
Article
256. Lahita RG, Kunkel HG, Bradlow HL. Increased oxidation of testosterone in systemic lupus erythematosus. Arthritis Rheum. 1983; 26:1517–1521.
Article
257. Lahita RG, Bradlow HL, Ginzler E, Pang S, New M. Low plasma androgens in women with systemic lupus erythematosus. Arthritis Rheum. 1987; 30:241–248.
Article
258. Tai P, Wang J, Jin H, Song X, Yan J, Kang Y, et al. Induction of regulatory T cells by physiological level estrogen. J Cell Physiol. 2008; 214:456–464.
Article
259. Peeva E, Venkatesh J, Diamond B. Tamoxifen blocks estrogen-induced B cell maturation but not survival. J Immunol. 2005; 175:1415–1423.
Article
Full Text Links
  • JRD
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr