DOI QR코드

DOI QR Code

Potential of Cells and Cytokines/Chemokines to Regulate Tertiary Lymphoid Structures in Human Diseases

  • Feifeng Jing (Department of Biomedical Sciences, University of Ulsan College of Medicine) ;
  • Eun Young Choi (Department of Biomedical Sciences, University of Ulsan College of Medicine)
  • Received : 2016.07.14
  • Accepted : 2016.08.27
  • Published : 2016.10.31

Abstract

Tertiary lymphoid structures (TLS) are ectopic lymphoid tissues involved in chronic inflammation, autoimmune diseases, transplant rejection and cancer. They exhibit almost all the characteristics of secondary lymphoid organs (SLO), which are associated with adaptive immune responses; as such, they contain organized B-cell follicles with germinal centers, distinct areas containing T cells and dendritic cells, high endothelial venules, and lymphatics. In this review, we briefly describe the formation of SLO, and describe the cellular subsets and molecular cues involved in the formation and maintenance of TLS. Finally, we discuss the associations of TLS with human diseases, especially autoimmune diseases, and the potential for therapeutic targeting.

Keywords

Acknowledgement

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology of Korea (2011-0014447).

References

  1. van de Pavert, S. A. and R. E. Mebius. 2010. New insights into the development of lymphoid tissues. Nat. Rev. Immunol. 10: 664-674. 
  2. Carragher, D. M., J. Rangel-Moreno, and T. D. Randall. 2008. Ectopic lymphoid tissues and local immunity. Semin. Immunol. 20: 26-42. 
  3. Joshi, N. S., E. H. kama-Garren, Y. Lu, D. Y. Lee, G. P. Chang, A. Li, M. DuPage, T. Tammela, N. R. Kerper, A. F. Farago, R. Robbins, D. M. Crowley, R. T. Bronson, and T. Jacks. 2015. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity 43: 579-590. 
  4. Stranford, S., and N. H. Ruddle. 2012. Follicular dendritic cells, conduits, lymphatic vessels, and high endothelial venules in tertiary lymphoid organs: Parallels with lymph node stroma. Front. Immunol. 3: 350. 
  5. Aloisi, F., and R. Pujol-Borrell. 2006. Lymphoid neogenesis in chronic inflammatory diseases. Nat. Rev. Immunol. 6: 205-217. 
  6. Buckley, C. D., F. Barone, S. Nayar, C. Benezech, and J. Caamano. 2015. Stromal cells in chronic inflammation and tertiary lymphoid organ formation. Annu. Rev. Immunol. 33: 715-745. 
  7. Humby, F., M. Bombardieri, A. Manzo, S. Kelly, M. C. Blades, B. Kirkham, J. Spencer, and C. Pitzalis. 2009. Ectopic lymphoid structures support ongoing production of class-switched autoantibodies in rheumatoid synovium. PLoS Med. 6: e1. 
  8. Thaunat, O., S. Graff-Dubois, S. Brouard, C. Gautreau, A. Varthaman, N. Fabien, A. C. Field, L. Louedec, J. Dai, E. Joly, E. Morelon, J. P. Soulillou, J. B. Michel, and A. Nicoletti. 2010. Immune responses elicited in tertiary lymphoid tissues display distinctive features. PLoS One 5: e11398. 
  9. Eberl, G. 2007. From induced to programmed lymphoid tissues: the long road to preempt pathogens. Trends Immunol. 28: 423-428. 
  10. Neyt, K., F. Perros, C. H. GeurtsvanKessel, H. Hammad, and B. N. Lambrecht. 2012. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol. 33: 297-305. 
  11. Pitzalis, C., G. W. Jones, M. Bombardieri, and S. A. Jones. 2014. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 14: 447-462. 
  12. Hsiao, H. M., W. Li, A. E. Gelman, A. S. Krupnick, and D. Kreisel. 2016. The role of lymphoid neogenesis in allografts. Am. J. Transplant. 16: 1079-1085. 
  13. Dieu-Nosjean, M. C., J. Goc, N. A. Giraldo, C. Sautes-Fridman, and W. H. Fridman. 2014. Tertiary lymphoid structures in cancer and beyond. Trends Immunol. 35: 571-580. 
  14. Drayton, D. L., S. Liao, R. H. Mounzer, and N. H. Ruddle. 2006. Lymphoid organ development: from ontogeny to neogenesis. Nat. Immunol. 7: 344-353. 
  15. Evans, I., and M. Y. Kim. 2009. Involvement of lymphoid inducer cells in the development of secondary and tertiary lymphoid structure. BMB Rep. 42: 189-193. 
  16. Marchesi, F., A. P. Martin, N. Thirunarayanan, E. Devany, L. Mayer, M. G. Grisotto, G. C. Furtado, and S. A. Lira. 2009. CXCL13 expression in the gut promotes accumulation of IL-22-producing lymphoid tissue-inducer cells, and formation of isolated lymphoid follicles. Mucosal Immunol. 2: 486-494. 
  17. Meier, D., C. Bornmann, S. Chappaz, S. Schmutz, L. A. Otten, R. Ceredig, H. cha-Orbea, and D. Finke. 2007. Ectopic lymphoid-organ development occurs through interleukin 7-mediated enhanced survival of lymphoid-tissue-inducer cells. Immunity 26: 643-654. 
  18. Kim, M. Y., F. M. Gaspal, H. E. Wiggett, F. M. McConnell, A. Gulbranson-Judge, C. Raykundalia, L. S. Walker, M. D. Goodall, and P. J. Lane. 2003. CD4(+)CD3(-) accessory cells costimulate primed CD4 T cells through OX40 and CD30 at sites where T cells collaborate with B cells. Immunity 18: 643-654. 
  19. Rangel-Moreno, J., J. E. Moyron-Quiroz, D. M. Carragher, K. Kusser, L. Hartson, A. Moquin, and T. D. Randall. 2009. Omental milky spots develop in the absence of lymphoid tissue-inducer cells and support B and T cell responses to peritoneal antigens. Immunity 30: 731-743. 
  20. Furtado, G. C., M. E. Pacer, G. Bongers, C. Benezech, Z. He, L. Chen, M. C. Berin, G. Kollias, J. H. Caamano, and S. A. Lira. 2014. TNFalpha-dependent development of lymphoid tissue in the absence of RORgammat(+) lymphoid tissue inducer cells. Mucosal Immunol. 7: 602-614. 
  21. Lochner, M., C. Ohnmacht, L. Presley, P. Bruhns, M. Si-Tahar, S. Sawa, and G. Eberl. 2011. Microbiota-induced tertiary lymphoid tissues aggravate inflammatory disease in the absence of RORgamma t and LTi cells. J. Exp. Med. 208: 125-134. 
  22. Peters, A., L. A. Pitcher, J. M. Sullivan, M. Mitsdoerffer, S. E. Acton, B. Franz, K. Wucherpfennig, S. Turley, M. C. Carroll, R. A. Sobel, E. Bettelli, and V. K. Kuchroo. 2011. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity 35: 986-996. 
  23. Guedj, K., J. Khallou-Laschet, M. Clement, M. Morvan, A. T. Gaston, G. Fornasa, J. Dai, M. Gervais-Taurel, G. Eberl, J. B. Michel, G. Caligiuri, and A. Nicoletti. 2014. M1 macrophages act as LTbetaR-independent lymphoid tissue inducer cells during atherosclerosis-related lymphoid neogenesis. Cardiovasc. Res. 101: 434-443. 
  24. Marinkovic, T., A. Garin, Y. Yokota, Y. X. Fu, N. H. Ruddle, G. C. Furtado, and S. A. Lira. 2006. Interaction of mature CD3+CD4+ T cells with dendritic cells triggers the development of tertiary lymphoid structures in the thyroid. J. Clin. Invest. 116: 2622-2632. 
  25. Okuda, M., A. Togawa, H. Wada, and S. Nishikawa. 2007. Distinct activities of stromal cells involved in the organogenesis of lymph nodes and Peyer's patches. J. Immunol. 179: 804-811. 
  26. Hu, D., S. K. Mohanta, C. Yin, L. Peng, Z. Ma, P. Srikakulapu, G. Grassia, N. MacRitchie, G. Dever, P. Gordon, F. L. Burton, A. Ialenti, S. R. Sabir, I. B. McInnes, J. M. Brewer, P. Garside, C. Weber, T. Lehmann, D. Teupser, L. Habenicht, M. Beer, R. Grabner, P. Maffia, F. Weih, and A. J. Habenicht. 2015. Artery tertiary lymphoid organs control aorta immunity and protect against atherosclerosis via vascular smooth muscle cell lymphotoxin beta receptors. Immunity 42: 1100-1115. 
  27. Kain, M. J., and B. M. Owens. 2013. Stromal cell regulation of homeostatic and inflammatory lymphoid organogenesis. Immunology 140: 12-21. 
  28. Pikor, N. B., A. Prat, A. Bar-Or, and J. L. Gommerman. 2015. Meningeal tertiary lymphoid tissues and multiple sclerosis: A gathering place for diverse types of immune cells during CNS autoimmunity. Front. Immunol. 6: 657. 
  29. Buckley, C. D. 2011. Why does chronic inflammation persist: An unexpected role for fibroblasts. Immunol. Lett. 138: 12-14. 
  30. Link, A., D. L. Hardie, S. Favre, M. R. Britschgi, D. H. Adams, M. Sixt, J. G. Cyster, C. D. Buckley, and S. A. Luther. 2011. Association of T-zone reticular networks and conduits with ectopic lymphoid tissues in mice and humans. Am. J. Pathol. 178: 1662-1675. 
  31. Nacionales, D. C., J. S. Weinstein, X. J. Yan, E. Albesiano, P. Y. Lee, K. M. Kelly-Scumpia, R. Lyons, M. Satoh, N. Chiorazzi, and W. H. Reeves. 2009. B cell proliferation, somatic hypermutation, class switch recombination, and autoantibody production in ectopic lymphoid tissue in murine lupus. J. Immunol. 182: 4226-4236. 
  32. Hansen, A., P. E. Lipsky, and T. Dorner. 2007. B cells in Sjogren's syndrome: indications for disturbed selection and differentiation in ectopic lymphoid tissue. Arthritis Res. Ther. 9: 218. 
  33. Weinstein, J. S., M. J. Delano, Y. Xu, K. M. Kelly-Scumpia, D. C. Nacionales, Y. Li, P. Y. Lee, P. O. Scumpia, L. Yang, E. Sobel, L. L. Moldawer, and W. H. Reeves. 2013. Maintenance of anti-Sm/RNP autoantibody production by plasma cells residing in ectopic lymphoid tissue and bone marrow memory B cells. J. Immunol. 190: 3916-3927. 
  34. McDonald, K. G., J. S. McDonough, and R. D. Newberry. 2005. Adaptive immune responses are dispensable for isolated lymphoid follicle formation: antigen-naive, lymphotoxin-sufficient B lymphocytes drive the formation of mature isolated lymphoid follicles. J. Immunol. 174: 5720-5728. 
  35. Dubey, L. K., L. Lebon, I. Mosconi, C. Y. Yang, E. Scandella, B. Ludewig, S. A. Luther, and N. L. Harris. 2016. Lymphotoxin-dependent B cell-FRC crosstalk promotes de novo follicle formation and antibody production following intestinal helminth infection. Cell Rep. 15: 1527-1541. 
  36. Dieu-Nosjean, M. C., N. A. Giraldo, H. Kaplon, C. Germain, W. H. Fridman, and C. Sautes-Fridman. 2016. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol. Rev. 271: 260-275. 
  37. Kroeger, D. R., K. Milne, and B. H. Nelson. 2016. Tumor-Infiltrating Plasma Cells Are Associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer. Clin. Cancer Res. 22: 3005-3015. 
  38. Kang, Y. M., X. Zhang, U. G. Wagner, H. Yang, R. D. Beckenbaugh, P. J. Kurtin, J. J. Goronzy, and C. M. Weyand. 2002. CD8 T cells are required for the formation of ectopic germinal centers in rheumatoid synovitis. J. Exp. Med. 195: 1325-1336. 
  39. Shinoda, K., K. Hirahara, T. Iinuma, T. Ichikawa, A. S. Suzuki, K. Sugaya, D. J. Tumes, H. Yamamoto, T. Hara, S. Tani-Ichi, K. Ikuta, Y. Okamoto, and T. Nakayama. 2016. Thy1+IL-7+lymphatic endothelial cells in iBALT provide a survival niche for memory T-helper cells in allergic airway inflammation. Proc. Natl. Acad. Sci. U. S. A. 113: E2842-E2851. 
  40. Benezech, C., N. T. Luu, J. A. Walker, A. A. Kruglov, Y. Loo, K. Nakamura, Y. Zhang, S. Nayar, L. H. Jones, A. Flores-Langarica, A. McIntosh, J. Marshall, F. Barone, G. Besra, K. Miles, J. E. Allen, M. Gray, G. Kollias, A. F. Cunningham, D. R. Withers, K. M. Toellner, N. D. Jones, M. Veldhoen, S. A. Nedospasov, A. N. McKenzie, and J. H. Caamano. 2015. Inflammation-induced formation of fat-associated lymphoid clusters. Nat. Immunol. 16: 819-828. 
  41. Sallusto, F., and A. Lanzavecchia. 2009. Human Th17 cells in infection and autoimmunity. Microbes Infect. 11: 620-624. 
  42. Grogan, J. L., and W. Ouyang. 2012. A role for Th17 cells in the regulation of tertiary lymphoid follicles. Eur. J. Immunol. 42: 2255-2262. 
  43. Pikor, N. B., J. L. Astarita, L. Summers-Deluca, G. Galicia, J. Qu, L. A. Ward, S. Armstrong, C. X. Dominguez, D. Malhotra, B. Heiden, R. Kay, V. Castanov, H. Touil, L. Boon, P. O'Connor, A. Bar-Or, A. Prat, V. Ramaglia, S. Ludwin, S. J. Turley, and J. L. Gommerman. 2015. Integration of Th17-and lymphotoxin-derived signals initiates meningeal-resident stromal cell remodeling to propagate neuroinflammation. Immunity 43: 1160-1173. 
  44. Deteix, C., V. ttuil-Audenis, A. Duthey, N. Patey, B. McGregor, V. Dubois, G. Caligiuri, S. Graff-Dubois, E. Morelon, and O. Thaunat. 2010. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J. Immunol. 184: 5344-5351. 
  45. Lu, K. T., Y. Kanno, J. L. Cannons, R. Handon, P. Bible, A. G. Elkahloun, S. M. Anderson, L. Wei, H. Sun, J. J. O'Shea, and P. L. Schwartzberg. 2011. Functional and epigenetic studies reveal multistep differentiation and plasticity of in vitro-generated and in vivo-derived follicular T helper cells. Immunity 35: 622-632. 
  46. Rangel-Moreno, J., D. M. Carragher, de la Luz Garcia-Hernandez, J. Y. Hwang, K. Kusser, L. Hartson, J. K. Kolls, S. A. Khader, and T. D. Randall. 2011. The development of inducible bronchus-associated lymphoid tissue depends on IL-17. Nat. Immunol. 12: 639-646. 
  47. Ma, C. S., and E. K. Deenick. 2014. Human T follicular helper (Tfh) cells and disease. Immunol. Cell Biol. 92: 64-71. 
  48. Romme, C. J., L. Bornsen, R. Ratzer, F. Piehl, M. Khademi, T. Olsson, P. S. Sorensen, and F. Sellebjerg. 2013. Systemic inflammation in progressive multiple sclerosis involves follicular T-helper, Th17- and activated B-cells and correlates with progression. PLoS One 8: e57820. 
  49. Vu, V. D., K. C. Beier, L. J. Pietzke, M. S. Al Baz, R. K. Feist, S. Gurka, E. Hamelmann, R. A. Kroczek, and A. Hutloff. 2016. Local T/B cooperation in inflamed tissues is supported by T follicular helper-like cells. Nat. Commun. 7: 10875. 
  50. Kocks, J. R., A. C. valos-Misslitz, G. Hintzen, L. Ohl, and R. Forster. 2007. Regulatory T cells interfere with the development of bronchus-associated lymphoid tissue. J. Exp. Med. 204: 723-734. 
  51. Foo, S. Y., V. Zhang, A. Lalwani, J. P. Lynch, A. Zhuang, C. E. Lam, P. S. Foster, C. King, R. J. Steptoe, S. B. Mazzone, P. D. Sly, and S. Phipps. 2015. Regulatory T cells prevent inducible BALT formation by dampening neutrophilic inflammation. J. Immunol. 194: 4567-4576. 
  52. Goc, J., W. H. Fridman, C. Sautes-Fridman, and M. C. eu-Nosjean. 2013. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology 2: e26836. 
  53. GeurtsvanKessel, C. H., M. A. Willart, I. M. Bergen, L. S. van Rijt, F. Muskens, D. Elewaut, A. D. Osterhaus, R. Hendriks, G. F. Rimmelzwaan, and B. N. Lambrecht. 2009. Dendritic cells are crucial for maintenance of tertiary lymphoid structures in the lung of influenza virus-infected mice. J. Exp. Med. 206: 2339-2349. 
  54. Halle, S., H. C. Dujardin, N. Bakocevic, H. Fleige, H. Danzer, S. Willenzon, Y. Suezer, G. Hammerling, N. Garbi, G. Sutter, T. Worbs, and R. Forster. 2009. Induced bronchus-associated lymphoid tissue serves as a general priming site for T cells and is maintained by dendritic cells. J. Exp. Med. 206: 2593-2601. 
  55. Park, C. S., and Y. S. Choi. 2005. How do follicular dendritic cells interact intimately with B cells in the germinal centre? Immunology 114: 2-10. 
  56. Nacionales, D. C., K. M. Kelly, P. Y. Lee, H. Zhuang, Y. Li, J. S. Weinstein, E. Sobel, Y. Kuroda, J. Akaogi, M. Satoh, and W. H. Reeves. 2006. Type I interferon production by tertiary lymphoid tissue developing in response to 2,6,10,14-tetramethyl-pentadecane (pristane). Am. J. Pathol. 168: 1227-1240. 
  57. Muniz, L. R., M. E. Pacer, S. A. Lira, and G. C. Furtado. 2011. A critical role for dendritic cells in the formation of lymphatic vessels within tertiary lymphoid structures. J. Immunol. 187: 828-834. 
  58. Goc, J., C. Germain, T. K. Vo-Bourgais, A. Lupo, C. Klein, S. Knockaert, C. L. de, H. Ouakrim, E. Becht, M. Alifano, P. Validire, R. Remark, S. A. Hammond, I. Cremer, D. Damotte, W. H. Fridman, C. Sautes-Fridman, and M. C. eu-Nosjean. 2014. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 74: 705-715. 
  59. Chiang, E. Y., G. A. Kolumam, X. Yu, M. Francesco, S. Ivelja, I. Peng, P. Gribling, J. Shu, W. P. Lee, C. J. Refino, M. Balazs, A. Paler-Martinez, A. Nguyen, J. Young, K. H. Barck, R. A. Carano, R. Ferrando, L. Diehl, D. Chatterjea, and J. L. Grogan. 2009. Targeted depletion of lymphotoxin-alpha-expressing TH1 and TH17 cells inhibits autoimmune disease. Nat. Med. 15: 766-773. 
  60. Griffith, J. W., C. L. Sokol, and A. D. Luster. 2014. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu. Rev. Immunol. 32: 659-702. 
  61. Weiss, J. M., M. Robinet, R. Aricha, P. Cufi, B. Villeret, F. Lantner, I. Shachar, S. Fuchs, M. C. Souroujon, S. Berrih-Aknin, and P. R. Le. 2016. Novel CXCL13 transgenic mouse: inflammation drives pathogenic effect of CXCL13 in experimental myasthenia gravis. Oncotarget 7: 7550-7562. 
  62. Ansel, K. M., V. N. Ngo, P. L. Hyman, S. A. Luther, R. Forster, J. D. Sedgwick, J. L. Browning, M. Lipp, and J. G. Cyster. 2000. A chemokine-driven positive feedback loop organizes lymphoid follicles. Nature 406: 309-314. 
  63. Summers-DeLuca, L. E., D. D. McCarthy, B. Cosovic, L. A. Ward, C. C. Lo, S. Scheu, K. Pfeffer, and J. L. Gommerman. 2007. Expression of lymphotoxin-alphabeta on antigen-specific T cells is required for DC function. J. Exp. Med. 204: 1071-1081. 
  64. Burman, A., O. Haworth, D. L. Hardie, E. N. Amft, C. Siewert, D. G. Jackson, M. Salmon, and C. D. Buckley. 2005. A chemokine-dependent stromal induction mechanism for aberrant lymphocyte accumulation and compromised lymphatic return in rheumatoid arthritis. J. Immunol. 174: 1693-1700. 
  65. Xu, X., Y. Han, Q. Wang, M. Cai, Y. Qian, X. Wang, H. Huang, L. Xu, L. Xiao, and B. Shi. 2016. Characterisation of tertiary lymphoid organs in explanted rejected donor kidneys. Immunol. Invest. 45: 38-51. 
  66. Ota, N., K. Wong, P. A. Valdez, Y. Zheng, N. K. Crellin, L. Diehl, and W. Ouyang. 2011. IL-22 bridges the lymphotoxin pathway with the maintenance of colonic lymphoid structures during infection with Citrobacter rodentium. Nat. Immunol. 12: 941-948. 
  67. Barone, F., S. Nayar, J. Campos, T. Cloake, D. R. Withers, K. M. Toellner, Y. Zhang, L. Fouser, B. Fisher, S. Bowman, J. Rangel-Moreno, M. L. Garcia-Hernandez, T. D. Randall, D. Lucchesi, M. Bombardieri, C. Pitzalis, S. A. Luther, and C. D. Buckley. 2015. IL-22 regulates lymphoid chemokine production and assembly of tertiary lymphoid organs. Proc. Natl. Acad. Sci. U. S. A .112: 11024-11029. 
  68. Canete, J. D., R. Celis, N. Yeremenko, R. Sanmarti, D. L. van, J. Ramirez, I. Blijdorp, C. M. Garcia-Herrero, J. L. Pablos, and D. L. Baeten. 2015. Ectopic lymphoid neogenesis is strongly associated with activation of the IL-23 pathway in rheumatoid synovitis. Arthritis Res. Ther. 17: 173. 
  69. Huang, H. Y., and S. A. Luther. 2012. Expression and function of interleukin-7 in secondary and tertiary lymphoid organs. Semin. Immunol. 24: 175-189. 
  70. Schmutz, S., N. Bosco, S. Chappaz, O. Boyman, H. cha-Orbea, R. Ceredig, A. G. Rolink, and D. Finke. 2009. Cutting edge: IL-7 regulates the peripheral pool of adult ROR gamma+lymphoid tissue inducer cells. J. Immunol. 183: 2217-2221. 
  71. Timmer, T. C., B. Baltus, M. Vondenhoff, T. W. Huizinga, P. P. Tak, C. L. Verweij, R. E. Mebius, and van der Pouw Kraan TC. 2007. Inflammation and ectopic lymphoid structures in rheumatoid arthritis synovial tissues dissected by genomics technology: identification of the interleukin-7 signaling pathway in tissues with lymphoid neogenesis. Arthritis Rheum. 56: 2492-2502. 
  72. Ciccia, F., G. Guggino, A. Rizzo, A. Manzo, B. Vitolo, M. P. La Manna, G. Giardina, G. Sireci, F. Dieli, C. M. Montecucco, R. Alessandro, and G. Triolo. 2015. Potential involvement of IL-9 and Th9 cells in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford) 54: 2264-2272. 
  73. Batten, M., N. Ramamoorthi, N. M. Kljavin, C. S. Ma, J. H. Cox, H. S. Dengler, D. M. Danilenko, P. Caplazi, M. Wong, D. A. Fulcher, M. C. Cook, C. King, S. G. Tangye, F. J. de Sauvage, and N. Ghilardi. 2010. IL-27 supports germinal center function by enhancing IL-21 production and the function of T follicular helper cells. J. Exp. Med. 207: 2895-2906. 
  74. Pickens, S. R., N. D. Chamberlain, M. V. Volin, A. M. Mandelin, H. Agrawal, M. Matsui, T. Yoshimoto, and S. Shahrara. 2011. Local expression of interleukin-27 ameliorates collagen-induced arthritis. Arthritis Rheum. 63: 2289-2298. 
  75. Manzo, A., M. Bombardieri, F. Humby, and C. Pitzalis. 2010. Secondary and ectopic lymphoid tissue responses in rheumatoid arthritis: from inflammation to autoimmunity and tissue damage/remodeling. Immunol. Rev. 233: 267-285. 
  76. Bombardieri, M., and C. Pitzalis. 2012. Ectopic lymphoid neogenesis and lymphoid chemokines in Sjogren's syndrome: at the interplay between chronic inflammation, autoimmunity and lymphomagenesis. Curr. Pharm. Biotechnol. 13: 1989-1996. 
  77. Chang, A., S. G. Henderson, D. Brandt, N. Liu, R. Guttikonda, C. Hsieh, N. Kaverina, T. O. Utset, S. M. Meehan, R. J. Quigg, E. Meffre, and M. R. Clark. 2011. In situ B cell-mediated immune responses and tubulointerstitial inflammation in human lupus nephritis. J. Immunol. 186: 1849-1860. 
  78. Kendall, P. L., G. Yu, E. J. Woodward, and J. W. Thomas. 2007. Tertiary lymphoid structures in the pancreas promote selection of B lymphocytes in autoimmune diabetes. J. Immunol. 178: 5643-5651. 
  79. Kielczewski, J. L., R. Horai, Y. Jittayasothorn, C. C. Chan, and R. R. Caspi. 2016. Tertiary lymphoid tissue forms in retinas of mice with spontaneous autoimmune uveitis and has consequences on visual function. J. Immunol. 196: 1013-1025. 
  80. Berrih-Aknin, S., S. Ragheb, P. R. Le, and R. P. Lisak. 2013. Ectopic germinal centers, BAFF and anti-B-cell therapy in myasthenia gravis. Autoimmun. Rev. 12: 885-893. 
  81. Risselada, A. P., M. F. Looije, A. A. Kruize, J. W. Bijlsma, and J. A. van Roon. 2013. The role of ectopic germinal centers in the immunopathology of primary Sjogren's syndrome: a systematic review. Semin. Arthritis Rheum. 42: 368-376. 
  82. Finkin, S., D. Yuan, I. Stein, K. Taniguchi, A. Weber, K. Unger, J. L. Browning, N. Goossens, S. Nakagawa, G. Gunasekaran, M. E. Schwartz, M. Kobayashi, H. Kumada, M. Berger, O. Pappo, K. Rajewsky, Y. Hoshida, M. Karin, M. Heikenwalder, Y. Ben-Neriah, and E. Pikarsky. 2015. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 16: 1235-1244. 
  83. Gregorio, A., C. Gambini, V. Gerloni, A. Parafioriti, M. P. Sormani, S. Gregorio, M. G. De, F. Rossi, A. Martini, and M. Gattorno. 2007. Lymphoid neogenesis in juvenile idiopathic arthritis correlates with ANA positivity and plasma cells infiltration. Rheumatology (Oxford) 46: 308-313. 
  84. Kuerten, S., A. Schickel, C. Kerkloh, M. S. Recks, K. Addicks, N. H. Ruddle, and P. V. Lehmann. 2012. Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response. Acta Neuropathol. 124: 861-873. 
  85. Motallebzadeh, R., S. Rehakova, T. M. Conlon, T. S. Win, C. J. Callaghan, M. Goddard, E. M. Bolton, N. H. Ruddle, J. A. Bradley, and G. J. Pettigrew. 2012. Blocking lymphotoxin signaling abrogates the development of ectopic lymphoid tissue within cardiac allografts and inhibits effector antibody responses. FASEB J. 26: 51-62. 
  86. Klimatcheva, E., T. Pandina, C. Reilly, S. Torno, H. Bussler, M. Scrivens, A. Jonason, C. Mallow, M. Doherty, M. Paris, E. S. Smith, and M. Zauderer. 2015. CXCL13 antibody for the treatment of autoimmune disorders. BMC Immunol. 16: 6. 
  87. Clement, M., K. Guedj, F. Andreata, M. Morvan, L. Bey, J. Khallou-Laschet, A. T. Gaston, S. Delbosc, J. M. Alsac, P. Bruneval, C. Deschildre, B. M. Le, Y. Castier, H. J. Kim, H. Cantor, J. B. Michel, G. Caligiuri, and A. Nicoletti. 2015. Control of the T follicular helper-germinal center B-cell axis by CD8(+) regulatory T cells limits atherosclerosis and tertiary lymphoid organ development. Circulation 131: 560-570. 
  88. Hamza, N., H. Bootsma, S. Yuvaraj, F. K. Spijkervet, E. A. Haacke, R. P. Pollard, A. Visser, A. Vissink, C. G. Kallenberg, F. G. Kroese, and N. A. Bos. 2012. Persistence of immunoglobulin-producing cells in parotid salivary glands of patients with primary Sjogren's syndrome after B cell depletion therapy. Ann. Rheum. Dis. 71: 1881-1887. 
  89. Kim, M. H., E. J. Taparowsky, and C. H. Kim. 2015. Retinoic Acid Differentially Regulates the Migration of Innate Lymphoid Cell Subsets to the Gut. Immunity 43: 107-119. 
  90. Ito, R., T. Takahashi, I. Katano, and M. Ito. 2012. Current advances in humanized mouse models. Cell. Mol. Immunol. 9: 208-214. 
  91. Croia, C., B. Serafini, M. Bombardieri, S. Kelly, F. Humby, M. Severa, F. Rizzo, E. M. Coccia, P. Migliorini, F. Aloisi, and C. Pitzalis. 2013. Epstein-Barr virus persistence and infection of autoreactive plasma cells in synovial lymphoid structures in rheumatoid arthritis. Ann. Rheum. Dis. 72: 1559-1568. 
  92. Grewal, J. S., M. J. Pilgrim, S. Grewal, L. Kasman, P. Werner, M. E. Bruorton, S. D. London, and L. London. 2011. Salivary glands act as mucosal inductive sites via the formation of ectopic germinal centers after site-restricted MCMV infection. FASEB J. 25: 1680-1696. 
  93. Kobayashi, Y., and T. Watanabe. 2010. Synthesis of artificial lymphoid tissue with immunological function. Trends Immunol. 31: 422-428.