Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Intercellular Protein Transfer from Thymocytes to Thymic Epithelial Cells

  • Hong-Xia Wang,

    Affiliations Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America, Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China

  • Yu-Rong Qiu ,

    xiaoping.zhong@duke.edu (XPZ); qiuyuronggz@126.com (YRQ)

    Affiliation Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China

  • Xiao-Ping Zhong

    xiaoping.zhong@duke.edu (XPZ); qiuyuronggz@126.com (YRQ)

    Affiliations Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America, Department of Immunology, Duke University Medical Center, Durham, NC, 27710, United States of America, Hematologic Malignancies and Cellular Therapies Program, Duke Cancer Institute, Duke University Medical Center, Durham, NC, 27710, United States of America

Abstract

Promiscuous expression of tissue restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs) is crucial for negative selection of self-reactive T cells to establish central tolerance. Intercellular transfer of self-peptide-MHC complexes from mTECs to thymic dendritic cells (DCs) allows DCs to acquire TRAs, which in turn contributes to negative selection and regulatory T cell generation. However, mTECs are unlikely to express all TRAs, such as immunoglobulins generated only in B cells after somatic recombination, hyper-mutation, or class-switches. We report here that both mTECs and cortical TECs can efficiently acquire not only cell surface but also intracellular proteins from thymocytes. This reveals a previously unappreciated intercellular sharing of molecules from thymocytes to TECs, which may broaden the TRA inventory in mTECs for establishing a full spectrum of central tolerance.

Introduction

Proper intrathymic T cell development ensures the generation of a repertoire of T cells against various pathogens but also self-tolerant. Thymus is composed of multiple cell lineages of different origins, such as developing T cells, dendritic cells (DCs), macrophages, B cells, and thymic epithelial cells (TECs). The thymus is separated into the cortex and medulla, which are involved in the distinct function of the thymus with regard to T cell development [13]. Early thymic progenitors enter the thymus at the conjunction between medulla and cortex. These cells, phenotypically CD4-CD8- double negative (DN), migrate toward the cortex to initiate early T cell development [4]. After successful recombination of the T cell receptor β gene and expression of the pre-TCRα/β receptor, these cells mature to the CD4+CD8+ double positive (DP) stage, at which the TCRα gene rearranges [5]. Expression of a functional αβ TCR on DP thymocytes and engagement of these TCRs with self-peptide major histocompatibility complex (MHC) expression on cortical TECs (cTECs) ensures their survival and differentiation to the CD4+CD8- and CD4-CD8+ single positive (SP) stage, also known as positive selection. SP thymocytes migrate into the medulla, where they engage with medullary TECs (mTECs) and DCs via TCR and self-peptide MHC interactions [1]. SP thymocytes expressing TCRs with high affinities to self-peptide–MHC complexes are self-reactive and are eliminated from the T cell repertoire due to programmed cell death, a process also called negative selection for establishing central tolerance. SP thymocytes with weak affinities to self-peptide–MHC complexes escape negative selection for populating peripheral lymphoid organs [6].

To establish central tolerance, mTECs must express tissue-restricted antigens (TRAs), which requires the transcription factor Aire [711]. Deficiency of Aire causes defective TRA expression, impaired mTECs maturation, and severe autoimmune diseases in both mice and humans [7, 12]. Besides directly triggering negative selection, mTECs share the burden with medullar DCs to establish central tolerance [13, 14]. Although DCs do not actively transcribe TRAs, they can acquire TRAs and self-peptide–MHC complexes from mTECs via intercellular protein transfer. Thymic DCs have been found to play important roles in negative selection of self-reactive T cells as well as for induction of regulatory T cells via acquisition of TRAs and MHCs from mTECs [1519]. Although Aire has the capacity to induce promiscuous transcription of TRAs in mTECs, it is hard to envision that all TRAs are actively transcribed in mTECs. Furthermore, some TRAs can only be generated after somatic recombination events that are strictly tissue/cell lineage specific, such as TCRs and immunoglobulins in thymocytes/T cells and B cells, respectively. Additional mechanisms must exist for mTECs and DCs to acquire TRAs. We report here that not only cell surface but also intracellular proteins can be efficiently transferred from thymocytes to both mTECs and cTECs, revealing a novel mechanism for mTECs to acquire thymocyte TRAs via intercellular transfer.

Materials and Methods

Ethics Statement

This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. Experiments in this study were performed according to protocols (A095-13-04) approved by the Institutional Animal Care and Usage Committee of Duke University.

Mice

C57BL6/J, Rosa26-LSL-ZsGreen [20], Rosa26-LSL-Tdtomato [20], and TCRα-/- [21] mice were purchased from the Jackson laboratory. CD4Cre mice [22] were purchased from Taconic Inc. Foxn1Cre mice [23] were kindly provided by Dr. Nancy Manley, University of Georgia. The mice were housed in a pathogen-free facility and were bred as described in the Results section. Mice were euthanized by CO2 followed by organ removal. Total 40 mice (18 male and 22 female mice) were used for experiments.

Antibodies and Flow Cytometry

The following antibodies used for flow cytometry were purchased from Biolegend: anti-CD4 (clone GK1.5), CD8 (clone 53–6.7), CD45 (clone 30-F11), CD45.2 (clone 104), EpCAM/CD326 (clone G8.8), Ly51 (clone 6C3), IgG isotype control, Ulex Europaeus Agglutinin I (UEA-1, clone B-1065; vector laboratories). Cells were stained for surface molecules using 2% FBS-PBS as previously described [24]. Cell death was identified by 7-AAD staining. Stained samples were acquired on a FACS Canto-II (BD Biosciences) flow cytometer. Data were analyzed with FlowJo software (Tree Star) and were gated on live cells and singlets.

Preparation of TEC single cell suspension

TEC single cell preparation was performed according to published protocols with modifications [25, 26]. Thymi were gently removed and trimmed of fat and connective tissue in cold RPMI-1640 containing 2% FBS. The thymus was then cut into small pieces (<2mm), which were suspended in 2ml digestion buffer containing 250μl collagenase type IV(10mg/ml; Worthington), 40μl DNase (50mg/ml; Worthington), and 1.7ml free-FBS RPMI-1640 shaking at 150–200 rpm at 37°C in an incubator for 8–10 min. Digested thymus remnants were settled at room temperature for 1 minute, and supernatants were transferred to new tubes. The remaining thymus fragments were digested three additional times. After digestion, combined samples were spun down at 472g for 5min. Cell pellets were resuspended in IMDM-10, washed two times by centrifugation, and eventually resuspended in cold EDTA/FACS buffer (5Mm EDTA, 2%FBS in PBS). Cells were immediately used for cell surface and intracellular staining with indicated antibodies.

Bone Marrow Chimeras

Bone marrow (BM) cells, isolated from Rosa-LSL-ZsGreen and Rosa-LSL-ZsGreen-CD4Cre donor mice, were depleted of T cells with a PE conjugated anti-CD3 antibody (Biolegend) and anti-PE microbeads (Miltenyi Biotec) according to the manufacturer’s protocol. C57BL/6J recipient mice were lethally irradiated (1000 rad) and intravenously injected with 15 x 106 BM cells. After reconstitution, mice were monitored for movement, fur color, and weight daily in the first two weeks and every other day afterwards. Mice with weight loss greater than 15% would be euthanized according to our approved protocol. All mice in these experiments were healthy prior to the experimental endpoint. One month after BM reconstitution, single cell suspensions from the thymus were prepared and stained for flow cytometry analysis.

Immunofluorescence microscopy

Thymus lobes were embedded in OCT (Leica Biosystems Richmond Inc.) and frozen immediately at -80°C. Frozen thin sections (5μm) were cut and fixed in a 1:1 mixture of acetone and methanol at -20°C for 8 minutes. Sections were air-dried and kept at -20°C. After being warmed up to room temperature (RT), the frozen sections were blocked with PBS containing 3% BSA and 0.1% Triton X-100 for 30–45 minutes at room temperature, stained with primary rat anti-mouse keratin 8 (KRT8, Troma-1, DSHB, University of Iowa; 1:50 dilution) or rabbit anti-mouse KRT5 (PRB-160P, Covance; 1:200 dilution), and finally stained with a secondary Rhodamine-conjugated donkey anti-rabbit IgG antibody (1:400 dilution) or Rhodamine-conjugated goat anti-rat IgG antibody (Jackson ImmunoResearch Laboratories Inc.; 1:300 dilution). After staining, samples were mounted with Vector mounting solution containing DAPI (Vector) and allowed to dry overnight at RT in the dark. Images were acquired using a Zeiss ApoTome Microscope and analyzed using Photoshop CS6 software.

Results

Expression of hematopoietic/thymocyte specific molecules in both cTECs and mTECs

Cells derived from hematopoietic stem cells express the protein tyrosine phosphatase CD45. In contrast, TECs are phenotypically defined as CD45-EpCAM+ [27]. When analyzing TECs from wild-type (WT) mice, we noted that the traditionally defined CD45-EpCAM+ TEC population actually positioned as CD45low (Fig 1A). To rule out the possibility that an intermediate level of CD45 expression detected in TECs by FACS analysis was due to autofluorescence of this population, we stained TECs simultaneously with a PECy7-labeled anti-CD45 and a FITC-labeled anti-CD45.2 or a FITC-labeled IgG isotype control. TECs, gated as CD45lowEpCAM+, were examined for CD45.2 or isotype control staining. As shown in Fig 1B, CD45.2 was detected in TECs at levels above isotype control staining, indicating that CD45 was indeed expressed on TECs, although it was expressed at levels lower than cells of the hematopoietic origin (CD45+EpCAM-). Further analysis revealed that both mTECs (UEA-1+Ly51-) and cTECs (UEA-1-Ly51+) expressed low levels of CD45.2 (Fig 1B). Moreover, other T cell specific markers, such as Thy1.2, CD4, and CD8, were also weakly detected in total TECs and m/cTECs. Together, these observations revealed that TECs appear to promiscuously express low levels of thymocytes/hematopoietic specific molecules.

thumbnail
Fig 1. Detection of thymocyte/hematopoietic specific cell surface markers in TECs.

A. Representative dot-plot of CD45 (PECy7) and EpCAM (APCCy7) staining of single cell suspension of thymus and UEA-1 (Biotin-strepavdin-APC) plus Ly51 (PE) staining in live and singlet gated CD45lowEpCAM+ TECs in WT thymocytes. B. Overlaid histogram for CD45.2 (FITC), Thy1.2 (FITC), CD4 (PECy5), and CD8 (FITC) or IgG isotype control staining in gated TECs (CD45lowEpCAM+), mTECs (CD45lowEpCAM+UEA-1+Ly51-), cTECs (CD45lowEpCAM+UEA-1-Ly51+), and CD45+EpCAM- thymocytes. Data shown are representative of at least three experiments.

https://doi.org/10.1371/journal.pone.0152641.g001

Transfer of intracellular fluorescent proteins from thymocytes to cTECs and mTECs

Low level expression of thymocyte-specific molecules in TECs, particularly in mTECs, could be the result of promiscuous transcription of these molecules in TECs. To rule out this possibility and to further examine whether TECs could acquire molecules inside thymocytes, we bred the Rosa-LSL-ZsGreen reporter mice [20] with mice carrying the CD4Cre transgene, which mediates T cell specific deletion of gene segments flanked by two loxp sites [22]. Deletion of the Loxp-STOP-Loxp cassette inserted upstream of the ZsGreen gene allows high level expression of ZsGreen protein in CD45+EpCAM- thymocyte (Fig 2A). More important, low level ZsGreen could also be detected in total TECs as well as cTECs and mTECs.

thumbnail
Fig 2. Intercellular transfer of intracellular fluorescent proteins from thymocytes to TECs.

A. Overlaid histogram for ZsGreen intensity in gated TECs (CD45.2lowEpCAM+), mTECs (CD45.2lowEpCAM+UEA-1+Ly51-), cTECs (CD45.2lowEpCAM+UEA-1-Ly51+), and CD45+EpCAM- and CD45.2+EpCAM- thymocytes from Rosa-LSL-ZsGreen-CD4Cre mice and Rosa-LSL-ZsGreen control mice. B. Overlaid histogram for ZsGreen intensity in gated TECs (CD45.2lowEpCAM+), mTECs (CD45.2lowEpCAM+UEA-1+Ly51-), and cTECs (CD45.2lowEpCAM+UEA-1-Ly51+), and CD45.2+EpCAM- thymocytes from Rosa-LSL-ZsGreen-Foxn1Cre mice and Rosa-LSL-ZsGreen control mice. C. Overlaid histogram for TdTomato intensity in gated TECs (CD45.2lowEpCAM+), mTECs (CD45.2lowEpCAM+UEA-1+Ly51-), and cTECs (CD45.2lowEpCAM+UEA-1-Ly51+), and CD45.2+EpCAM- thymocytes from Rosa-LSL-TdTomato-CD4Cre mice and Rosa-LSL-TdTomato control mice. D. Detection of ZsGreen in TECs from irradiation chimeric C57BL/6J mice reconstituted with BM cells from Rosa-LSL-ZsGreen-CD4Cre mice. C57BL6/J mice were lethally irradiated (1000 rad) and reconstituted with BM cells from Rosa-LSL-ZsGreen-CD4Cre mice or Rosa-LSL-ZsGreen control mice. Thirty days after transfer, TECs from recipients were examined for ZsGreen intensity. Overlaid histogram shows ZsGreen intensity in gated mTECs (CD45.2lowEpCAM+UEA-1+Ly51-), cTECs (CD45.2lowEpCAM+UEA-1-Ly51+), and CD45.2+EpCAM- thymocytes from recipient mice. E. ZsGreen+ thymocytes were unable to transfer ZsGreen to TECs during ex vivo TEC preparation. Total thymocytes from Rosa-LSL-ZsGreen-CD4Cre mice were pelleted and resuspended in 50 μl digestion buffer and transferred to Rosa-LSL-ZsGreen thymus during TEC preparation, starting at the tearing of the thymus. The single cell suspension was similarly stained as in Fig 1. Overlaid histograms show ZsGreen intensity in TECs from Rosa-LSL-ZsGreen thymus, whether or not mixed with ZsGreen+ thymocytes. TECs from Rosa-LSL-ZsGreen-CD4Cre mice were used as positive controls. Data shown are representative of three (A–D) and two (E) experiments with at least one pair of mice in each experiment.

https://doi.org/10.1371/journal.pone.0152641.g002

To rule out that low levels of ZsGreen expression in the TECs were not caused by Cre-mediated deletion of the Loxp-STOP-Loxp cassette in these cells, we examined the same Rosa-LSL-ZsGreen reporter mice carrying the Foxn1Cre transgene [23], which mediates TEC-specific deletion of the Loxp-STOP-Loxp cassette. ZsGreen in TECs from Rosa-LSL-ZsGreen-Foxn1Cre mice was directly under the control of the Rasa26 promoter, and the actin promoter knocked into the locus. As shown in Fig 2B, ZsGreen level in these TECs was much higher than that detected in Rosa-LSL-ZsGreen-CD4Cre mice. Such thymocyte to TEC transfer of intracellular proteins was not limited to ZsGreen. TECs from CD4Cre mice carrying a conditional Tdtomato reporter, Rosa26-LSL-TdTommato [20], were also detected to weakly express Tdtomato (Fig 2C). Together, these observations revealed that intracellular proteins in the thymocytes can be transferred into TECs.

Although data from Rosa-LSL-ZsGreen-CD4Cre mice supported that intercellular transfer of proteins from thymocytes to TECs occurred in the thymus, they did not firmly rule out that active transcription and translation of low levels of ZsGreen in TECs in Rosa-LSL-ZsGreen-CD4Cre mice. To address this issue, we generated irradiation chimeric mice using BM cells from Rosa-LSL-ZsGreen-CD4Cre mice. One month after reconstitution, we detected not only high levels of ZsGreen expression in the thymocytes, but also significant ZsGreen expression in both mTECs and cTECs (Fig 2D), ruling out that ZsGreen expression in TECs in these mice was resulted from low levels of ZsGreen transcription and translation in these cells.

To further rule out that the ZsGreen detected in TECs from Rosa-LSL-ZsGreen-CD4Cre mice by flow-cytometry was an artifact introduced during ex vivo processing of the thymus such as formation of TEC—thymocyte conjugates or uptake of Zsgreen protein released from thymocytes, we first isolated total thymocytes from Rosa-LSL-ZsGreen-CD4Cre mice and then mixed these ZsGreen+ thymocytes in high density with Rosa-LSL-ZsGreen thymus during TEC preparation. As shown in Fig 2E, ZsGreen intensity in TECs from the preparation mixed with ZsGreen+ thymocytes was similar to that in TECs without mixed with ZsGreen+ thymocytes, suggesting that ex vivo preparation of TECs was not sufficient to transfer ZsGreen from thymocytes to TECs.

Impact of TCR-MHC engagement on protein transfer from thymocytes to TECs

Developing thymocytes engage with self-peptide–MHC complexes expressed on cTECs and mTECs via TCRs to mediated positive and negative selection, respectively. To examine whether such engagement may affect protein transfer from the thymocytes to TECs, we analyzed TCRα-/-ZsGreeen-CD4Cre and TCRα+/-ZsGreeen-CD4Cre mice. Similar to previously reported [21], SP thymocytes were virtually absent in TCRα-/-ZsGreeen-CD4Cre mice, but not in TCRα+/-ZsGreeen-CD4Cre mice. Crosstalk between SP thymocytes and TECs is important for mTEC maturation and survival [2832]. TEC percentages and total TEC numbers were decreased in TCRα-/-ZsGreeen-CD4Cre mice (Fig 3A and 3B), accompanying obvious decreases of mTEC percentages and numbers (Fig 3C–3E). Although cTEC percentages were relatively increased, cTEC numbers were not noticeably changed in TCRα-/-ZsGreeen-CD4Cre mice compared with TCRα+/-ZsGreeen-CD4Cre mice. Importantly, ZsGreen intensity in both mTECs and cTECs from TCRα-/-ZsGreeen-CD4Cre mice were lower than those from TCRα+/-ZsGreeen-CD4Cre mice (Fig 3F and 3G). Because DP thymocytes from both mice expressed similar levels of ZsGreen (Fig 3H), it suggested that decreased ZsGreeen intensity in TCRα-/-ZsGreeen-CD4Cre TECs was not caused by abnormal ZsGreen expression in thymocytes from these mice. Thus, engagement between TECs and thymocytes via TCR-MHC interactions might positively contributed to protein transfer from thymocytes to TECs. Alternatively, TCR-MHC interaction could promote protein uptake by mTECs through promoting mTEC maturation. Because Zsgreen levels in TECs from TCRα-/-ZsGreeen-CD4Cre mice were still higher than those in TECs from ZsGreen negative control mice, this data suggested that TCR-MHC independent mechanism(s) also contributed to protein transfer from thymocytes to TECs.

thumbnail
Fig 3. Contribution of TCR-MHC engagement dependent and independent mechanisms to protein transfer from thymocytes to TECs.

A, B. Frequencies (A) and absolute numbers (B) of TECs in TCRα-/-ZsGreeen-CD4Cre and TCRα+/-ZsGreeen-CD4Cre mice. C. Representative dot plots of showing mTEC and cTEC subsets. D, E. Percentages (D) and absolute numbers (E) of mTECs and cTEC. F. Overlaid histograms showing ZsGreen level in mTECs and cTECs. G. Relative gMFI of ZsGreen in m/cTECs. gMFI in TCRα+/-ZsGreeenf/+CD4Cre TECs was arbitrarily set as 1. H. ZsGreen intensity of CD45+EpCAM-CD4+CD8+ DP thymocytes. Data shown are representative or calculated from three experiments. *, p<0.05 determined by Student t-test. Raw data for Fig 3A, 3B, 3D, 3E, and 3G are shown in S1 Data.

https://doi.org/10.1371/journal.pone.0152641.g003

Discussion

Here, we provide the first evidence that mTECs and cTECs are capable of acquisition of both cell surface and intracellular proteins from thymocytes. Because TCRα deficiency considerably decreased ZsGreen acquisition by TECs from thymocytes, TCR-MHC engagement-dependent mechanisms must be involved in intercellular protein transfer from thymocytes to TECs. However, we cannot rule out whether third parties such as DCs and macrophages are also involved in the transfer from thymocyte to TECs. Additionally, our data suggests that TCR independent transfer of protein from thymocyte to TECs occurs. At present, how proteins are transferred from thymocytes to TECs remains unknown, but the process is likely involved in multiple mechanisms. Trogocytosis, which transfers membrane patches and associated proteins through “membrane nibbling,” nanotubes connecting different cells, and gap junctions have been reported to be involved in intercellular protein transfer [3335]. Thymocytes can also release exomes [36, 37], which could be uptaken by TECs. Additionally, apoptotic thymocytes could be uptaken by DCs and macrophages or directly by TECs as well as through release of thymocyte specific components in local environment and subsequently uptaken by TECs. In support of this possibility, we have found that TECs are able to uptake soluble proteins in vitro and process uptaken proteins in an acidic compartment (Data not shown).

Previous reports have demonstrated intercellular transfer of proteins from mTECs to thymic DCs [13, 14]. Unidirectional transfer of self-peptide–MHC complexes from mTECs to DCs allows DCs to present self-TRAs to SP thymocytes for induction of negative selection and generation of regulatory T cells[1519]. Although the physiological importance of protein transfer from thymocytes to TECs remains to be illuminated, it is conceivable that such transfer may broaden TRA inventory in mTECs by inclusion of antigens that are normally only expressed in thymocytes but not transcribed in mTECs. For example, TCRs are only generated after somatic V(D)J recombination that occurs strictly in thymocytes. Transfer of TCRs and other T cell specific proteins from thymocytes to TECs may provide a pathway to allow T cell–specific antigens to be presented by TECs, particularly mTECs, which may induce negative selection of self-reactive T cells against T cell–restricted antigens such as TCRs. Interestingly, a recent study has found that intrathymic B cells are licensed to present TRAs to induce negative selection [38]. It would be interesting to determine whether thymic B cells may uniquely present immunoglobulin epitopes to prevent generation of B cell–reactive T cells.

Supporting Information

S1 Data. Raw data for analyses shown in Fig 3 of the manuscript.

https://doi.org/10.1371/journal.pone.0152641.s001

(XLSX)

Acknowledgments

We thank Dr. Nancy Manley for kindly providing us the Foxn1Cre mice.

Author Contributions

Conceived and designed the experiments: HXW YRQ XPZ. Performed the experiments: HXW. Analyzed the data: HXW YRQ XPZ. Wrote the paper: HXW XPZ.

References

  1. 1. Anderson G, Takahama Y. Thymic epithelial cells: working class heroes for T cell development and repertoire selection. Trends Immunol. 2012;33(6):256–63. Epub 2012/05/18. pmid:22591984.
  2. 2. Rodewald H-R. Thymus organogenesis. Annu Rev Immunol. 2008;26(18304000):355–88.
  3. 3. Manley NR, Richie ER, Blackburn CC, Condie BG, Sage J. Structure and function of the thymic microenvironment. Frontiers in bioscience: a journal and virtual library. 2011;16:2461–77. Epub 2011/05/31. pmid:21622189.
  4. 4. Yang Q, Jeremiah Bell J, Bhandoola A. T-cell lineage determination. Immunol Rev. 2010;238(1):12–22. pmid:20969581; PubMed Central PMCID: PMC2972740.
  5. 5. Krangel MS, Hernandez-Munain C, Lauzurica P, McMurry M, Roberts JL, Zhong XP. Developmental regulation of V(D)J recombination at the TCR alpha/delta locus. Immunol Rev. 1998;165:131–47. Epub 1998/12/16. pmid:9850858.
  6. 6. Klein L, Kyewski B, Allen PM, Hogquist KA. Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see). Nat Rev Immunol. 2014;14(6):377–91. pmid:24830344.
  7. 7. Peterson P, Org T, Rebane A. Transcriptional regulation by AIRE: molecular mechanisms of central tolerance. Nat Rev Immunol. 2008;8(12):948–57. Epub 2008/11/15. pmid:19008896; PubMed Central PMCID: PMC2785478.
  8. 8. Anderson MS, Su MA. Aire and T cell development. Curr Opin Immunol. 2011;23(2):198–206. Epub 2010/12/18. pmid:21163636; PubMed Central PMCID: PMC3073725.
  9. 9. Roberts NA, White AJ, Jenkinson WE, Turchinovich G, Nakamura K, Withers DR, et al. Rank signaling links the development of invariant gammadelta T cell progenitors and Aire(+) medullary epithelium. Immunity. 2012;36(3):427–37. pmid:22425250; PubMed Central PMCID: PMC3368267.
  10. 10. Coder BD, Wang H, Ruan L, Su DM. Thymic involution perturbs negative selection leading to autoreactive T cells that induce chronic inflammation. J Immunol. 2015;194(12):5825–37. pmid:25957168; PubMed Central PMCID: PMC4458423.
  11. 11. Mathis D, Benoist C. Aire. Annu Rev Immunol. 2009;27:287–312. Epub 2009/03/24. pmid:19302042.
  12. 12. Yano M, Kuroda N, Han H, Meguro-Horike M, Nishikawa Y, Kiyonari H, et al. Aire controls the differentiation program of thymic epithelial cells in the medulla for the establishment of self-tolerance. J Exp Med. 2008;205(12):2827–38. pmid:19015306; PubMed Central PMCID: PMC2585853.
  13. 13. Lopes N, Serge A, Ferrier P, Irla M. Thymic Crosstalk Coordinates Medulla Organization and T-Cell Tolerance Induction. Front Immunol. 2015;6:365. Epub 2015/08/11. pmid:26257733; PubMed Central PMCID: PMC4507079.
  14. 14. Klein L, Hinterberger M, von Rohrscheidt J, Aichinger M. Autonomous versus dendritic cell-dependent contributions of medullary thymic epithelial cells to central tolerance. Trends Immunol. 2011;32(5):188–93. Epub 2011/04/16. pmid:21493141.
  15. 15. Koble C, Kyewski B. The thymic medulla: a unique microenvironment for intercellular self-antigen transfer. J Exp Med. 2009;206(7):1505–13. Epub 2009/07/01. pmid:19564355; PubMed Central PMCID: PMC2715082.
  16. 16. Hubert FX, Kinkel SA, Davey GM, Phipson B, Mueller SN, Liston A, et al. Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood. 2011;118(9):2462–72. Epub 2011/04/21. pmid:21505196.
  17. 17. Gallegos AM, Bevan MJ. Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med. 2004;200(8):1039–49. Epub 2004/10/20. pmid:15492126; PubMed Central PMCID: PMC2211843.
  18. 18. Millet V, Naquet P, Guinamard RR. Intercellular MHC transfer between thymic epithelial and dendritic cells. Eur J Immunol. 2008;38(5):1257–63. Epub 2008/04/17. pmid:18412162.
  19. 19. Perry JS, Lio CW, Kau AL, Nutsch K, Yang Z, Gordon JI, et al. Distinct contributions of Aire and antigen-presenting-cell subsets to the generation of self-tolerance in the thymus. Immunity. 2014;41(3):414–26. pmid:25220213; PubMed Central PMCID: PMC4175925.
  20. 20. Madisen L, Zwingman TA, Sunkin SM, Oh SW, Zariwala HA, Gu H, et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci. 2010;13(1):133–40. Epub 2009/12/22. pmid:20023653; PubMed Central PMCID: PMC2840225.
  21. 21. Mombaerts P, Clarke AR, Rudnicki MA, Iacomini J, Itohara S, Lafaille JJ, et al. Mutations in T-cell antigen receptor genes alpha and beta block thymocyte development at different stages. Nature. 1992;360(6401):225–31. Epub 1992/11/19. pmid:1359428.
  22. 22. Lee PP, Fitzpatrick DR, Beard C, Jessup HK, Lehar S, Makar KW, et al. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity. 2001;15(5):763–74. Epub 2001/12/01. pmid:11728338.
  23. 23. Gordon J, Xiao S, Hughes B, Su D-m, Navarre SP, Condie BG, et al. Specific expression of lacZ and cre recombinase in fetal thymic epithelial cells by multiplex gene targeting at the Foxn1 locus. BMC Dev Biol. 2007;7(17577402):69-.
  24. 24. Guo R, Wan CK, Carpenter JH, Mousallem T, Boustany RM, Kuan CT, et al. Synergistic control of T cell development and tumor suppression by diacylglycerol kinase alpha and zeta. Proc Natl Acad Sci U S A. 2008;105(33):11909–14. pmid:18689679; PubMed Central PMCID: PMC2575297.
  25. 25. Gray DHD, Fletcher AL, Hammett M, Seach N, Ueno T, Young LF, et al. Unbiased analysis, enrichment and purification of thymic stromal cells. J Immunol Methods. 2008;329(17988680):56–66.
  26. 26. Wang HX, Shin J, Wang S, Gorentla B, Lin X, Gao J, et al. mTORC1 in Thymic Epithelial Cells Is Critical for Thymopoiesis, T-Cell Generation, and Temporal Control of γδT17 Development and TCRγ/δ Recombination. PLoS Biol. 2016;14(2):e1002370. pmid:26889835; PubMed Central PMCID: PMC4758703.
  27. 27. Manley NR, Blackburn CC. A developmental look at thymus organogenesis: where do the non-hematopoietic cells in the thymus come from? Curr Opin Immunol. 2003;15(2):225–32. Epub 2003/03/14. pmid:12633674.
  28. 28. Palmer DB, Viney JL, Ritter MA, Hayday AC, Owen MJ. Expression of the alpha beta T-cell receptor is necessary for the generation of the thymic medulla. Dev Immunol. 1993;3(3):175–9. pmid:8281032; PubMed Central PMCID: PMC2275932.
  29. 29. Yang SJ, Ahn S, Park CS, Holmes KL, Westrup J, Chang CH, et al. The quantitative assessment of MHC II on thymic epithelium: implications in cortical thymocyte development. Int Immunol. 2006;18(5):729–39. pmid:16569676.
  30. 30. Irla M, Hugues S, Gill J, Nitta T, Hikosaka Y, Williams IR, et al. Autoantigen-specific interactions with CD4+ thymocytes control mature medullary thymic epithelial cell cellularity. Immunity. 2008;29(18799151):451–63.
  31. 31. Shores EW, Van Ewijk W, Singer A. Disorganization and restoration of thymic medullary epithelial cells in T cell receptor-negative scid mice: evidence that receptor-bearing lymphocytes influence maturation of the thymic microenvironment. Eur J Immunol. 1991;21(7):1657–61. pmid:2060577.
  32. 32. Surh CD, Ernst B, Sprent J. Growth of epithelial cells in the thymic medulla is under the control of mature T cells. J Exp Med. 1992;176(2):611–6. pmid:1500862; PubMed Central PMCID: PMC2119324.
  33. 33. Neijssen J, Herberts C, Drijfhout JW, Reits E, Janssen L, Neefjes J. Cross-presentation by intercellular peptide transfer through gap junctions. Nature. 2005;434(7029):83–8. Epub 2005/03/04. pmid:15744304.
  34. 34. Rustom A, Saffrich R, Markovic I, Walther P, Gerdes HH. Nanotubular highways for intercellular organelle transport. Science. 2004;303(5660):1007–10. Epub 2004/02/14. pmid:14963329.
  35. 35. Onfelt B, Nedvetzki S, Yanagi K, Davis DM. Cutting edge: Membrane nanotubes connect immune cells. J Immunol. 2004;173(3):1511–3. Epub 2004/07/22. pmid:15265877.
  36. 36. Skogberg G, Lundberg V, Berglund M, Gudmundsdottir J, Telemo E, Lindgren S, et al. Human thymic epithelial primary cells produce exosomes carrying tissue-restricted antigens. Immunol Cell Biol. 2015. Epub 2015/03/18. pmid:25776846.
  37. 37. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79. Epub 2002/08/03. pmid:12154376.
  38. 38. Yamano T, Nedjic J, Hinterberger M, Steinert M, Koser S, Pinto S, et al. Thymic B Cells Are Licensed to Present Self Antigens for Central T Cell Tolerance Induction. Immunity. 2015;42(6):1048–61. Epub 2015/06/14. pmid:26070482.