Select the scenarios that describe negative selection. select the two answers that are correct.

If you're seeing this message, it means we're having trouble loading external resources on our website.

If you're behind a web filter, please make sure that the domains *.kastatic.org and *.kasandbox.org are unblocked.

1. Kyewski B, Klein L. A central role for central tolerance. Annual review of immunology. 2006;24:571–606. [PubMed] [Google Scholar]

2. Nakagawa Y, Ohigashi I, Nitta T, Sakata M, Tanaka K, et al. Thymic nurse cells provide microenvironment for secondary T cell receptor alpha rearrangement in cortical thymocytes. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:20572–20577. [PMC free article] [PubMed] [Google Scholar]

3. Klein L, Hinterberger M, Wirnsberger G, Kyewski B. Antigen presentation in the thymus for positive selection and central tolerance induction. Nature reviews. Immunology. 2009;9:833–844. [PubMed] [Google Scholar]

4. Florea BI, Verdoes M, Li N, van der Linden WA, Geurink PP, et al. Activity-based profiling reveals reactivity of the murine thymoproteasome-specific subunit beta5t. Chemistry & biology. 2010;17:795–801. [PMC free article] [PubMed] [Google Scholar]

5. Murata S, Sasaki K, Kishimoto T, Niwa S, Hayashi H, et al. Regulation of CD8+ T cell development by thymus-specific proteasomes. Science. 2007;316:1349–1353. [PubMed] [Google Scholar]

6. Nakagawa T, Roth W, Wong P, Nelson A, Farr A, et al. Cathepsin L: critical role in Ii degradation and CD4 T cell selection in the thymus. Science. 1998;280:450–453. [PubMed] [Google Scholar]

7. Gommeaux J, Gregoire C, Nguessan P, Richelme M, Malissen M, et al. Thymus-specific serine protease regulates positive selection of a subset of CD4+ thymocytes. European journal of immunology. 2009;39:956–964. [PubMed] [Google Scholar]

8. Nedjic J, Aichinger M, Mizushima N, Klein L. Macroautophagy, endogenous MHC II loading and T cell selection: the benefits of breaking the rules. Current opinion in immunology. 2009;21:92–97. [PubMed] [Google Scholar]

9. Nedjic J, Aichinger M, Emmerich J, Mizushima N, Klein L. Autophagy in thymic epithelium shapes the T-cell repertoire and is essential for tolerance. Nature. 2008;455:396–400. [PubMed] [Google Scholar]

10. Honey K, Nakagawa T, Peters C, Rudensky A. Cathepsin L regulates CD4+ T cell selection independently of its effect on invariant chain: a role in the generation of positively selecting peptide ligands. The Journal of experimental medicine. 2002;195:1349–1358. [PMC free article] [PubMed] [Google Scholar]

11. Nitta T, Murata S, Sasaki K, Fujii H, Ripen AM, et al. Thymoproteasome shapes immunocompetent repertoire of CD8+ T cells. Immunity. 2010;32:29–40. [PubMed] [Google Scholar]

12. Xing Y, Jameson SC, Hogquist KA. Thymoproteasome subunit-beta5T generates peptide-MHC complexes specialized for positive selection. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:6979–6984. [PMC free article] [PubMed] [Google Scholar]

13. Ziegler A, Muller CA, Bockmann RA, Uchanska-Ziegler B. Low-affinity peptides and T-cell selection. Trends in immunology. 2009;30:53–60. [PubMed] [Google Scholar]

14. Ryan KR, McNeil LK, Dao C, Jensen PE, Evavold BD. Modification of peptide interaction with MHC creates TCR partial agonists. Cellular immunology. 2004;227:70–78. [PubMed] [Google Scholar]

15. Azzam HS, Grinberg A, Lui K, Shen H, Shores EW, et al. CD5 expression is developmentally regulated by T cell receptor (TCR) signals and TCR avidity. Journal of Experimental Medicine. 1998;188:2301–2311. [PMC free article] [PubMed] [Google Scholar]

16. Stefanova I, Dorfman JR, Germain RN. Self-recognition promotes the foreign antigen sensitivity of naive T lymphocytes. Nature. 2002;420:429–434. [PubMed] [Google Scholar]

17. Cho JH, Kim HO, Surh CD, Sprent J. T cell receptor-dependent regulation of lipid rafts controls naive CD8+ T cell homeostasis. Immunity. 2010;32:214–226. [PMC free article] [PubMed] [Google Scholar]

18. Palmer MJ, Mahajan VS, Chen J, Irvine DJ, Lauffenburger DA. Signaling thresholds govern heterogeneity in IL-7-receptor-mediated responses of naive CD8(+) T cells. Immunology and cell biology. 2011;89:581–594. [PMC free article] [PubMed] [Google Scholar]

19. Mandl JN, Monteiro JP, Vrisekoop N, Germain RN. T cell-positive selection uses self-ligand binding strength to optimize repertoire recognition of foreign antigens. Immunity. 2013;38:263–274. [PMC free article] [PubMed] [Google Scholar]

20. Persaud SP, Parker CR, Lo WL, Weber KS, Allen PM. Intrinsic CD4(+) T cell sensitivity and response to a pathogen are set and sustained by avidity for thymic and peripheral complexes of self peptide and MHC. Nat Immunol. 2014;15:266–274.
[Refs. 18 and 19 find that T cell responsiveness is set in the thymus and maintained in mature T cells in proportion to the avidity of the positively selecting interaction. Ref. 18 concludes that T cells with stronger affinity for self dominate in responses to infections, whereas Ref. 19 challenges the generality of such correlations.] [PMC free article] [PubMed] [Google Scholar]

21. Surh CD, Sprent J. T-cell apoptosis detected in situ during positive and negative selection in the thymus. Nature. 1994;372:100–103. [PubMed] [Google Scholar]

22. Daley SR, Hu DY, Goodnow CC. Helios marks strongly autoreactive CD4+ T cells in two major waves of thymic deletion distinguished by induction of PD-1 or NF-kappaB. The Journal of experimental medicine. 2013;210:269–285. [PMC free article] [PubMed] [Google Scholar]

23. Stritesky GL, Xing Y, Erickson JR, Kalekar LA, Wang X, et al. Murine thymic selection quantified using a unique method to capture deleted T cells. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:4679–4684.
[Using different approaches, these two studies quantify ‘early’ and ‘late’ negative selection in the cortex and the medulla, respectively, and conclude that the extent of clonal deletion in the cortex exceeds that in the medulla.] [PMC free article] [PubMed] [Google Scholar]

24. McCaughtry TM, Baldwin TA, Wilken MS, Hogquist KA. Clonal deletion of thymocytes can occur in the cortex with no involvement of the medulla. The Journal of experimental medicine. 2008;205:2575–2584. [PMC free article] [PubMed] [Google Scholar]

25. Melichar HJ, Ross JO, Herzmark P, Hogquist KA, Robey EA. Distinct temporal patterns of T cell receptor signaling during positive versus negative selection in situ. Science signaling. 2013;6:ra92. [PMC free article] [PubMed] [Google Scholar]

26. Irla M, Hollander G, Reith W. Control of central self-tolerance induction by autoreactive CD4+ thymocytes. Trends in immunology. 2010;31:71–79. [PubMed] [Google Scholar]

27. Mathis D, Benoist C. Aire. Annual review of immunology. 2009;27:287–312. [PubMed] [Google Scholar]

28. Peterson P, Org T, Rebane A. Transcriptional regulation by AIRE: molecular mechanisms of central tolerance. Nature reviews. Immunology. 2008;8:948–957. [PMC free article] [PubMed] [Google Scholar]

29. Gallegos AM, Bevan MJ. Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. Journal of Experimental Medicine. 2004;200:1039–1049. [PMC free article] [PubMed] [Google Scholar]

30. Oukka M, Cohen-Tannoudji M, Tanaka Y, Babinet C, Kosmatopoulos K. Medullary thymic epithelial cells induce tolerance to intracellular proteins. Journal of immunology. 1996;156:968–975. [PubMed] [Google Scholar]

31. Hinterberger M, Aichinger M, Prazeres da Costa O, Voehringer D, Hoffmann R, et al. Autonomous role of medullary thymic epithelial cells in central CD4(+) T cell tolerance. Nature immunology. 2010;11:512–519.
[Through diminution of MHC class II on mTECs, this study documents an autonomous contribution of mTECs to both dominant and recessive mechanisms of CD4+ T cell tolerance and provides experimental support for the avidity model of TReg cell development versus clonal deletion.] [PubMed] [Google Scholar]

32. Klein L, Klein T, Ruther U, Kyewski B. CD4 T cell tolerance to human C-reactive protein, an inducible serum protein, is mediated by medullary thymic epithelium. The Journal of experimental medicine. 1998;188:5–16. [PMC free article] [PubMed] [Google Scholar]

33. Oukka M, Colucci-Guyon E, Tran PL, Cohen-Tannoudji M, Babinet C, et al. CD4 T cell tolerance to nuclear proteins induced by medullary thymic epithelium. Immunity. 1996;4:545–553. [PubMed] [Google Scholar]

34. Aschenbrenner K, D'Cruz LM, Vollmann EH, Hinterberger M, Emmerich J, et al. Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nature immunology. 2007;8:351–358. [PubMed] [Google Scholar]

35. Atibalentja DF, Byersdorfer CA, Unanue ER. Thymus-blood protein interactions are highly effective in negative selection and regulatory T cell induction. Journal of immunology. 2009;183:7909–7918. [PMC free article] [PubMed] [Google Scholar]

36. Klein L, Roettinger B, Kyewski B. Sampling of complementing self-antigen pools by thymic stromal cells maximizes the scope of central T cell tolerance. European journal of immunology. 2001;31:2476–2486. [PubMed] [Google Scholar]

37. Munz C. Enhancing immunity through autophagy. Annual review of immunology. 2009;27:423–449. [PubMed] [Google Scholar]

38. Aichinger M, Wu C, Nedjic J, Klein L. Macroautophagy substrates are loaded onto MHC class II of medullary thymic epithelial cells for central tolerance. The Journal of experimental medicine. 2013;210:287–300. [PMC free article] [PubMed] [Google Scholar]

39. Mizushima N. Autophagy in protein and organelle turnover. Cold Spring Harbor symposia on quantitative biology. 2011;76:397–402. [PubMed] [Google Scholar]

40. Dongre AR, Kovats S, deRoos P, McCormack AL, Nakagawa T, et al. In vivo MHC class II presentation of cytosolic proteins revealed by rapid automated tandem mass spectrometry and functional analyses. European journal of immunology. 2001;31:1485–1494. [PubMed] [Google Scholar]

41. Klein L, Hinterberger M, von Rohrscheidt J, Aichinger M. Autonomous versus dendritic cell-dependent contributions of medullary thymic epithelial cells to central tolerance. Trends in immunology. 2011;32:188–193. [PubMed] [Google Scholar]

42. Koble C, Kyewski B. The thymic medulla: a unique microenvironment for intercellular self-antigen transfer. The Journal of experimental medicine. 2009;206:1505–1513. [PMC free article] [PubMed] [Google Scholar]

43. Hubert FX, Kinkel SA, Davey GM, Phipson B, Mueller SN, et al. Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood. 2011;118:2462–2472. [PubMed] [Google Scholar]

44. Taniguchi RT, DeVoss JJ, Moon JJ, Sidney J, Sette A, et al. Detection of an autoreactive T-cell population within the polyclonal repertoire that undergoes distinct autoimmune regulator (Aire)-mediated selection. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:7847–7852. [PMC free article] [PubMed] [Google Scholar]

45. Irla M, Hugues S, Gill J, Nitta T, Hikosaka Y, et al. Autoantigen-specific interactions with CD4+ thymocytes control mature medullary thymic epithelial cell cellularity. Immunity. 2008;29:451–463. [PubMed] [Google Scholar]

46. DeVoss J, Hou Y, Johannes K, Lu W, Liou GI, et al. Spontaneous autoimmunity prevented by thymic expression of a single self-antigen. The Journal of experimental medicine. 2006;203:2727–2735. [PMC free article] [PubMed] [Google Scholar]

47. Fan Y, Rudert WA, Grupillo M, He J, Sisino G, et al. Thymus-specific deletion of insulin induces autoimmune diabetes. The EMBO journal. 2009;28:2812–2824. [PMC free article] [PubMed] [Google Scholar]

48. Ehrlich LI, Oh DY, Weissman IL, Lewis RS. Differential contribution of chemotaxis and substrate restriction to segregation of immature and mature thymocytes. Immunity. 2009;31:986–998. [PMC free article] [PubMed] [Google Scholar]

49. Le Borgne M, Ladi E, Dzhagalov I, Herzmark P, Liao YF, et al. The impact of negative selection on thymocyte migration in the medulla. Nature immunology. 2009;10:823–830. [PMC free article] [PubMed] [Google Scholar]

50. Ueda Y, Katagiri K, Tomiyama T, Yasuda K, Habiro K, et al. Mst1 regulates integrin-dependent thymocyte trafficking and antigen recognition in the thymus. Nature communications. 2012;3:1098. [PubMed] [Google Scholar]

51. Klein L. Dead man walking: how thymocytes scan the medulla. Nature immunology. 2009;10:809–811. [PubMed] [Google Scholar]

52. Derbinski J, Pinto S, Rosch S, Hexel K, Kyewski B. Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:657–662. [PMC free article] [PubMed] [Google Scholar]

53. Pinto S, Michel C, Schmidt-Glenewinkel H, Harder N, Rohr K, et al. Overlapping gene coexpression patterns in human medullary thymic epithelial cells generate self-antigen diversity. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:E3497–3505. [PMC free article] [PubMed] [Google Scholar]

54. Villasenor J, Besse W, Benoist C, Mathis D. Ectopic expression of peripheral-tissue antigens in the thymic epithelium: probabilistic, monoallelic, misinitiated. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:15854–15859. [PMC free article] [PubMed] [Google Scholar]

55. Wu L, Shortman K. Heterogeneity of thymic dendritic cells. Seminars in immunology. 2005;17:304–312. [PubMed] [Google Scholar]

56. Li J, Park J, Foss D, Goldschneider I. Thymus-homing peripheral dendritic cells constitute two of the three major subsets of dendritic cells in the steady-state thymus. The Journal of experimental medicine. 2009;206:607–622. [PMC free article] [PubMed] [Google Scholar]

57. Joffre OP, Segura E, Savina A, Amigorena S. Cross-presentation by dendritic cells. Nature reviews. Immunology. 2012;12:557–569. [PubMed] [Google Scholar]

58. Proietto AI, Lahoud MH, Wu L. Distinct functional capacities of mouse thymic and splenic dendritic cell populations (vol 86, pg 700, 2007). Immunology and Cell Biology. 2009;87:190–190. [PubMed] [Google Scholar]

59. Lei Y, Ripen AM, Ishimaru N, Ohigashi I, Nagasawa T, et al. Aire-dependent production of XCL1 mediates medullary accumulation of thymic dendritic cells and contributes to regulatory T cell development. The Journal of experimental medicine. 2011;208:383–394. [PMC free article] [PubMed] [Google Scholar]

60. Baba T, Nakamoto Y, Mukaida N. Crucial contribution of thymic Sirp alpha+ conventional dendritic cells to central tolerance against blood-borne antigens in a CCR2-dependent manner. Journal of immunology. 2009;183:3053–3063. [PubMed] [Google Scholar]

61. Atibalentja DF, Murphy KM, Unanue ER. Functional redundancy between thymic CD8alpha+ and Sirpalpha+ conventional dendritic cells in presentation of blood-derived lysozyme by MHC class II proteins. Journal of immunology. 2011;186:1421–1431. [PMC free article] [PubMed] [Google Scholar]

62. Baba T, Badr Mel S, Tomaru U, Ishizu A, Mukaida N. Novel process of intrathymic tumor-immune tolerance through CCR2-mediated recruitment of Sirpalpha+ dendritic cells: a murine model. PloS one. 2012;7:e41154. [PMC free article] [PubMed] [Google Scholar]

63. Reizis B, Colonna M, Trinchieri G, Barrat F, Gilliet M. Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system? Nature reviews. Immunology. 2011;11:558–565. [PMC free article] [PubMed] [Google Scholar]

64. Villadangos JA, Young L. Antigen-presentation properties of plasmacytoid dendritic cells. Immunity. 2008;29:352–361. [PubMed] [Google Scholar]

65. Wirnsberger G, Mair F, Klein L. Regulatory T cell differentiation of thymocytes does not require a dedicated antigen-presenting cell but is under T cell-intrinsic developmental control. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:10278–10283. [PMC free article] [PubMed] [Google Scholar]

66. Hadeiba H, Lahl K, Edalati A, Oderup C, Habtezion A, et al. Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance. Immunity. 2012;36:438–450.
[This study shows that endogenous pDCs take up subcutaneously injected antigen and transport it to the thymus in a CCR9-dependent fashion. Upon intravenous injection, antigen-loaded pDCs deleted specific thymocytes, revealing that migratory pDCs can support central tolerance.] [PMC free article] [PubMed] [Google Scholar]

67. Hadeiba H, Sato T, Habtezion A, Oderup C, Pan J, et al. CCR9 expression defines tolerogenic plasmacytoid dendritic cells able to suppress acute graft-versus-host disease. Nature immunology. 2008;9:1253–1260. [PMC free article] [PubMed] [Google Scholar]

68. Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, et al. Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nature immunology. 2006;7:1092–1100. [PubMed] [Google Scholar]

69. Akashi K, Richie LI, Miyamoto T, Carr WH, Weissman IL. B lymphopoiesis in the thymus. Journal of immunology. 2000;164:5221–5226. [PubMed] [Google Scholar]

70. Feyerabend TB, Terszowski G, Tietz A, Blum C, Luche H, et al. Deletion of Notch1 converts pro-T cells to dendritic cells and promotes thymic B cells by cell-extrinsic and cell-intrinsic mechanisms. Immunity. 2009;30:67–79. [PubMed] [Google Scholar]

71. Mori S, Inaba M, Sugihara A, Taketani S, Doi H, et al. Presence of B cell progenitors in the thymus. Journal of immunology. 1997;158:4193–4199. [PubMed] [Google Scholar]

72. Perera J, Meng L, Meng F, Huang H. Autoreactive thymic B cells are efficient antigen-presenting cells of cognate self-antigens for T cell negative selection. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:17011–17016.
[Using B cell receptor (BCR) and TCR transgenic mice, this study shows that autoreactive thymic B cells serve as efficient APCs for negative selection. Thymic B cells may capture autoantigens through their BCR and present these to developing thymocytes for clonal deletion.] [PMC free article] [PubMed] [Google Scholar]

73. Frommer F, Waisman A. B cells participate in thymic negative selection of murine auto-reactive CD4+ T cells. PloS one. 2010;5:e15372. [PMC free article] [PubMed] [Google Scholar]

74. Kleindienst P, Chretien I, Winkler T, Brocker T. Functional comparison of thymic B cells and dendritic cells in vivo. Blood. 2000;95:2610–2616. [PubMed] [Google Scholar]

75. Guerri L, Peguillet I, Geraldo Y, Nabti S, Premel V, et al. Analysis of APC types involved in CD4 tolerance and regulatory T cell generation using reaggregated thymic organ cultures. Journal of immunology. 2013;190:2102–2110. [PubMed] [Google Scholar]

76. Yuseff MI, Pierobon P, Reversat A, Lennon-Dumenil AM. How B cells capture, process and present antigens: a crucial role for cell polarity. Nature reviews. Immunology. 2013;13:475–486. [PubMed] [Google Scholar]

77. Weiss S, Bogen B. MHC class II-restricted presentation of intracellular antigen. Cell. 1991;64:767–776. [PubMed] [Google Scholar]

78. Munthe LA, Corthay A, Os A, Zangani M, Bogen B. Systemic autoimmune disease caused by autoreactive B cells that receive chronic help from Ig V region-specific T cells. Journal of immunology. 2005;175:2391–2400. [PubMed] [Google Scholar]

79. Detanico T, Heiser RA, Aviszus K, Bonorino C, Wysocki LJ. Self-tolerance checkpoints in CD4 T cells specific for a peptide derived from the B cell antigen receptor. Journal of immunology. 2011;187:82–91. [PMC free article] [PubMed] [Google Scholar]

80. Ebert PJ, Jiang S, Xie J, Li QJ, Davis MM. An endogenous positively selecting peptide enhances mature T cell responses and becomes an autoantigen in the absence of microRNA miR-181a. Nature immunology. 2009;10:1162–1169. [PMC free article] [PubMed] [Google Scholar]

81. Lo WL, Felix NJ, Walters JJ, Rohrs H, Gross ML, et al. An endogenous peptide positively selects and augments the activation and survival of peripheral CD4+ T cells. Nature immunology. 2009;10:1155–1161. [PMC free article] [PubMed] [Google Scholar]

82. Martin B, Auffray C, Delpoux A, Pommier A, Durand A, et al. Highly self-reactive naive CD4 T cells are prone to differentiate into regulatory T cells. Nature communications. 2013;4:2209. [PubMed] [Google Scholar]

83. Hsieh CS, Lee HM, Lio CW. Selection of regulatory T cells in the thymus. Nature reviews. Immunology. 2012;12:157–167. [PubMed] [Google Scholar]

84. Wirnsberger G, Hinterberger M, Klein L. Regulatory T-cell differentiation versus clonal deletion of autoreactive thymocytes. Immunology and cell biology. 2011;89:45–53. [PubMed] [Google Scholar]

85. Cowan JE, Parnell SM, Nakamura K, Caamano JH, Lane PJ, et al. The thymic medulla is required for Foxp3+ regulatory but not conventional CD4+ thymocyte development. The Journal of experimental medicine. 2013;210:675–681. [PMC free article] [PubMed] [Google Scholar]

86. Klein L, Jovanovic K. Regulatory T cell lineage commitment in the thymus. Seminars in immunology. 2011;23:401–409. [PubMed] [Google Scholar]

87. Mathis D, Benoist C. A decade of AIRE. Nature reviews. Immunology. 2007;7:645–650. [PubMed] [Google Scholar]

88. Malchow S, Leventhal DS, Nishi S, Fischer BI, Shen L, et al. Aire-dependent thymic development of tumor-associated regulatory T cells. Science. 2013;339:1219–1224.
[This study reports that TReg cells recurrently enriched in prostate tumors of mice recognized an unknown antigen that was also present in the healthy prostate. These cells were found to differentiate as ‘natural’ (i.e. thymically induced) TReg cells in an Aire-dependent manner, providing evidence for a link between Aire-mediated expression of peripheral tissue antigens and the development of organ-specific TReg cells.] [PMC free article] [PubMed] [Google Scholar]

89. Bautista JL, Lio CW, Lathrop SK, Forbush K, Liang Y, et al. Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nature immunology. 2009;10:610–617. [PMC free article] [PubMed] [Google Scholar]

90. Leung MW, Shen S, Lafaille JJ. TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes. The Journal of experimental medicine. 2009;206:2121–2130. [PMC free article] [PubMed] [Google Scholar]

91. Moran AE, Holzapfel KL, Xing Y, Cunningham NR, Maltzman JS, et al. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. The Journal of experimental medicine. 2011;208:1279–1289. [PMC free article] [PubMed] [Google Scholar]

92. St-Pierre C, Brochu S, Vanegas JR, Dumont-Lagace M, Lemieux S, et al. Transcriptome sequencing of neonatal thymic epithelial cells. Scientific reports. 2013;3:1860. [PMC free article] [PubMed] [Google Scholar]

93. Lv H, Havari E, Pinto S, Gottumukkala RV, Cornivelli L, et al. Impaired thymic tolerance to alpha-myosin directs autoimmunity to the heart in mice and humans. The Journal of clinical investigation. 2011;121:1561–1573. [PMC free article] [PubMed] [Google Scholar]

94. Gottumukkala RV, Lv H, Cornivelli L, Wagers AJ, Kwong RY, et al. Myocardial infarction triggers chronic cardiac autoimmunity in type 1 diabetes. Science translational medicine. 2012;4:138ra180. [PMC free article] [PubMed] [Google Scholar]

95. Gotter J, Brors B, Hergenhahn M, Kyewski B. Medullary epithelial cells of the human thymus express a highly diverse selection of tissue-specific genes colocalized in chromosomal clusters. The Journal of experimental medicine. 2004;199:155–166. [PMC free article] [PubMed] [Google Scholar]

96. Durinovic-Bello I, Wu RP, Gersuk VH, Sanda S, Shilling HG, et al. Insulin gene VNTR genotype associates with frequency and phenotype of the autoimmune response to proinsulin. Genes and immunity. 2010;11:188–193. [PMC free article] [PubMed] [Google Scholar]

97. Pugliese A, Zeller M, Fernandez A, Jr., Zalcberg LJ, Bartlett RJ, et al. The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nature genetics. 1997;15:293–297. [PubMed] [Google Scholar]

98. Vafiadis P, Bennett ST, Todd JA, Nadeau J, Grabs R, et al. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nature genetics. 1997;15:289–292. [PubMed] [Google Scholar]

99. Giraud M, Taubert R, Vandiedonck C, Ke X, Levi-Strauss M, et al. An IRF8-binding promoter variant and AIRE control CHRNA1 promiscuous expression in thymus. Nature. 2007;448:934–937. [PubMed] [Google Scholar]

100. Colobran R, Armengol Mdel P, Faner R, Gartner M, Tykocinski LO, et al. Association of an SNP with intrathymic transcription of TSHR and Graves’ disease: a role for defective thymic tolerance. Human molecular genetics. 2011;20:3415–3423. [PubMed] [Google Scholar]

101. Klein L, Klugmann M, Nave KA, Tuohy VK, Kyewski B. Shaping of the autoreactive T-cell repertoire by a splice variant of self protein expressed in thymic epithelial cells. Nature medicine. 2000;6:56–61. [PubMed] [Google Scholar]

102. de Jong VM, Abreu JR, Verrijn Stuart AA, van der Slik AR, Verhaeghen K, et al. Alternative splicing and differential expression of the islet autoantigen IGRP between pancreas and thymus contributes to immunogenicity of pancreatic islets but not diabetogenicity in humans. Diabetologia. 2013;56:2651–2658. [PubMed] [Google Scholar]

103. Scally SW, Petersen J, Law SC, Dudek NL, Nel HJ, et al. A molecular basis for the association of the HLA-DRB1 locus, citrullination, and rheumatoid arthritis. The Journal of experimental medicine. 2013 [PMC free article] [PubMed] [Google Scholar]

104. van Lummel M, Duinkerken G, van Veelen PA, de Ru A, Cordfunke R, et al. Post-Translational Modification Of Hla-Dq Binding Islet-Autoantigens In Type 1 Diabetes. Diabetes. 2013 [PubMed] [Google Scholar]

105. Gascoigne NR, Palmer E. Signaling in thymic selection. Current opinion in immunology. 2011;23:207–212. [PMC free article] [PubMed] [Google Scholar]

106. Bains I, van Santen HM, Seddon B, Yates AJ. Models of self-peptide sampling by developing T cells identify candidate mechanisms of thymic selection. PLoS Comput Biol. 2013;9:e1003102. [PMC free article] [PubMed] [Google Scholar]

107. Org T, Chignola F, Hetenyi C, Gaetani M, Rebane A, et al. The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression. EMBO reports. 2008;9:370–376. [PMC free article] [PubMed] [Google Scholar]

108. Koh AS, Kuo AJ, Park SY, Cheung P, Abramson J, et al. Aire employs a histone-binding module to mediate immunological tolerance, linking chromatin regulation with organ-specific autoimmunity. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:15878–15883. [PMC free article] [PubMed] [Google Scholar]

109. Abramson J, Giraud M, Benoist C, Mathis D. Aire's partners in the molecular control of immunological tolerance. Cell. 2010;140:123–135. [PubMed] [Google Scholar]

110. Giraud M, Yoshida H, Abramson J, Rahl PB, Young RA, et al. Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:535–540. [PMC free article] [PubMed] [Google Scholar]

111. Danso-Abeam D, Humblet-Baron S, Dooley J, Liston A. Models of aire-dependent gene regulation for thymic negative selection. Frontiers in immunology. 2011;2:14. [PMC free article] [PubMed] [Google Scholar]

112. Marrack P, Ignatowicz L, Kappler JW, Boymel J, Freed JH. Comparison of peptides bound to spleen and thymus class II. The Journal of experimental medicine. 1993;178:2173–2183. [PMC free article] [PubMed] [Google Scholar]

113. Collado JA, Alvarez I, Ciudad MT, Espinosa G, Canals F, et al. Composition of the HLA-DR-associated human thymus peptidome. European journal of immunology. 2013 [PubMed] [Google Scholar]

114. Espinosa G, Collado JA, Scholz E, Mestre-Ferrer A, Kuse N, et al. Peptides presented by HLA class I molecules in the human thymus. Journal of proteomics. 2013;94C:23–36. [PubMed] [Google Scholar]

115. Adamopoulou E, Tenzer S, Hillen N, Klug P, Rota IA, et al. Exploring the MHC-peptide matrix of central tolerance in the human thymus. Nature communications. 2013;4:2039. [PubMed] [Google Scholar]

116. Fortier MH, Caron E, Hardy MP, Voisin G, Lemieux S, et al. The MHC class I peptide repertoire is molded by the transcriptome. The Journal of experimental medicine. 2008;205:595–610. [PMC free article] [PubMed] [Google Scholar]

117. Mester G, Hoffmann V, Stevanovic S. Insights into MHC class I antigen processing gained from large-scale analysis of class I ligands. Cellular and molecular life sciences : CMLS. 2011;68:1521–1532. [PubMed] [Google Scholar]

118. Millet V, Naquet P, Guinamard RR. Intercellular MHC transfer between thymic epithelial and dendritic cells. European journal of immunology. 2008;38:1257–1263. [PubMed] [Google Scholar]


Page 2

Select the scenarios that describe negative selection. select the two answers that are correct.

Stromal cell interactions during T cell development

(a) Successive stages of double-negative (DN) T cell development are accompanied by an outward movement of thymocytes towards the sub-capsular zone. Subsequent to β-selection at the DN3 stage, double-positive (DP) cells ‘randomly walk’ through the outer cortex, which possibly facilitates the ‘scanning’ of cortical thymic epithelial cells (cTECs) for positively selecting ligands. At this stage, DP thymocytes may be engulfed by cTECs and form so-called thymic nurse cells (TNCs), whereby the molecular control and physiological relevance of this process remains to be established. Interactions of DP cells with cortical conventional dendritic cells (cDCs) may lead to negative selection. It remains open whether these cortical cDCs exclusively belong to the migratory Sirpα+ subset. Positively selected, CD4 or CD8 lineage-committed thymocytes relocate into the medulla by directed migration. Upon reaching the medulla, single-positive (SP) cells again assume a ‘random walk’ motion pattern. Through this random migration, SP cells may now ‘scan’ resident (res.) and migratory (migr.) cDCs, medullary thymic epithelial cells (mTECs), plasmacytoid dendritic cells (pDCs) and B cells. It is estimated that SP cells engage in around five contacts with antigen presenting cells (APCs) per hour, so that over their 4-5 days residency in the medulla, T cells may serially interact with several hundred APCs. (b) Key functional properties of thymic APCs discussed in this Review.

  • Select the scenarios that describe negative selection. select the two answers that are correct.
  • Select the scenarios that describe negative selection. select the two answers that are correct.
  • Select the scenarios that describe negative selection. select the two answers that are correct.
  • Select the scenarios that describe negative selection. select the two answers that are correct.

Click on the image to see a larger version.