Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jun 1.
Published in final edited form as: Curr Opin Immunol. 2019 May 28;58:83–88. doi: 10.1016/j.coi.2019.04.010

Development, ontogeny and maintenance of TCRαβ+ CD8αα IEL

Roland Ruscher 1,*, Kristin A Hogquist 1,#
PMCID: PMC6612447  NIHMSID: NIHMS1530365  PMID: 31146182

Abstract

The intestinal epithelium is the outermost cellular layer that separates the body from the gut lumen. The integrity of this protective mucosal barrier is crucial and maintained by specialized cells—intraepithelial lymphocytes (IEL). Much research has been conducted on these cells and our overall understanding of them is increasing rapidly. In this review we focus on the TCRαβ+ subset of CD8αα IEL. We discuss recent studies that shed light on the development, ontogeny, maintenance and functional characteristics of CD8αα IEL, and highlight yet unanswered questions for future studies.

Introduction

Intestinal intraepithelial lymphocytes (IEL) are comprised of innate lymphoid cells and T cells within the gut epithelium [13]. The T cell IEL include conventional resident memory TCRαβ+ CD4+ and CD8αβ+ T cells, as well as unconventional TCRαβ+CD4CD8αα+β and TCRγδ+ IEL [1,4]. Despite these differences in their origin, IEL bear notable functional and phenotypic commonality, including expression of T-bet, NK receptors of the Ly49 family, IL-15 receptors, and an activated phenotype with high expression of CD69 [1,5]. IEL patrol the epithelium and help to maintain homeostasis and integrity of the barrier surface between gut lumen and the body [1,6,7]. Effector mechanisms include stimulation of antimicrobial peptide production in epithelial cells through HVEM stimulation [8] and epithelial cell proliferation via an AHR regulated pathway [9]. IEL further have a cytolytic profile, including Granzyme B expression [1,4,10]. Within IEL, this review will focus on the TCRαβ+CD4 CD8αα+β IEL (hereafter called CD8αα IEL) and emerging concepts about their development.

Intestinal CD8αα IEL

The two main features that distinguish CD8αα IEL from conventional CD8+ or CD4+ IEL are: 1) self-reactive TCRs and 2) a co-receptor that consists solely of CD8αα homodimers. CD8αα homodimers bind the nonclassical MHC class I molecule thymus leukemia antigen (TL), which is expressed by intestinal epithelial cells [11,12]. Some research suggests CD8αα regulates TCR signaling strength, and TL-deficient mice develop more severe disease in experimental models of inflammatory bowel disease (IBD) [11,13]. CD8αα homodimer expression is also driven by distinct control elements at the Cd8a locus [14], although what upstream signals regulate it has not been fully flushed out. Regarding TCR self-reactivity in IEL, even though no specific self-peptide ligands for CD8αα IEL have yet been identified, several lines of evidence support the idea that this population is self-reactive. First, superantigen reactive “forbidden” Vβs are present amongst IEL [15]. Second, in TCR transgenic systems, CD8αα IEL numbers are increased by agonist self peptides [16]. Third, CD8αα IEL and their precursors are increased in Bim-deficient and in Bcl-XL transgenic mice, in which normally deleted self-reactive cells are rescued [1719]. Lastly, the thymic precursors for CD8αα IEL (IELp) have an activated phenotype and express high levels of Nur77GFP [18,19]. Interestingly, hydrophobic amino acids such as cysteine at certain positions within the complementary determining region 3 (CDR3) of the TCR have recently been suggested to promote self-reactivity in cells such as IELp [20,21].

Regarding the MHC restriction of these cells, it has long been known that gut CD8αα are strongly reduced in number in β2M deficient mice that lack expression of all classical and non-classical MHC class I molecules [19,22,23]. However the abundance of thymic IELp was only mildly reduced by β2M deficiency [19], suggesting that class I molecules (including TL) may have a role in the tissue maintenance of CD8αα IEL, as well as act as a restricting element for TCR specificity and selection in the thymus. Of CD8αα IEL TCRs characterized to date, many are restricted to nonclassical MHC class-I molecules, but clones restricted to classical MHC class-I and MHC class-II were also found [18,24], and MHC-crossreactivity was common amongst thymic CD8αα IEL precursors [25].

It should be noted that the overwhelming majority of research on CD8αα IEL has been conducted in mice. A recent report however indicates that a similar subset might also exist in humans, albeit ‘masked’ by CD8αβ coreceptor expression in addition to the CD8αα homodimer [26], which should spur more research on these cells in humans in the future.

Thymic development of CD8αα IEL precursors

Recent work has shed much light on the developmental pathways of CD8αα IEL. Although there was controversy in the past about the site of CD8αα IEL development [15,27,28], there now is agreement that the majority of these cells arise in the thymus [1,4]. IELp have been identified within the CD4CD8 (DN) TCRαβ+ pool [1719,29,30]. During thymic maturation, IELp, like conventional T cells, upregulate MHC class-I (i.e. H-2Kb in C57BL/6 mice) and also CD122 [18,19,31]. CD122 is a shared subunit of the IL-15 receptor, a cytokine that is essential for CD8αα IEL survival. Nonetheless, thymic IELp do not depend on IL-15 [19,30,32], implying that the relevant source of IL-15 is in the gut [33].

IELp have high expression of CD69, Nur77GFP, PD-1, and Egr2, indicative of strong and recent TCR activation[18,19]. But how do IELp escape deletion when they are strongly signaled in the thymus? Interestingly, IELp numbers were greatly enhanced in Bim-deficient mice, wherein strongly TCR-signaled thymocytes escape apoptosis [19,34,35]. These results suggest a model where strongly signaled thymocytes are sometimes deleted and sometimes give rise to CD8αα IEL. Indeed, retrogenic expression of TCRs cloned from CD8αα IEL supported both clonal deletion and IEL generation in the thymus, with an estimated event outcome of 97:3, respectively [18]. This fate decision could be stochastic as has been suggested for self-reactive regulatory T cells (Treg) [36,37] or directed by non-TCR factors, a process referred to as “clonal diversion”. In this context, the presence or absence of CD28 costimulation was shown to influence the fate of strongly signaled self-reactive thymocytes: While CD80/86-CD28 signaling normally results in deletion of self-reactive T cells in the thymus, the absence of CD80/86 or CD28 induced differentiation into the CD8αα IEL pathway and increased gut CD8αα IEL numbers [23]. Further, recent work showed that PD-1 inhibits CD28 signaling [38,39], suggesting the possibility that PD-1 facilitates IELp survival and differentiation by blocking CD28 signaling. Nonetheless, this remains to be experimentally proven, and the relevant APC that express CD80 and CD86, which presumably reside in the thymic cortex, have not been identified. An absence of CD28 signaling may not be the only factor favoring IEL differentiation. Like Treg and invariant natural killer T (iNKT) cells, IELp selection is dependent on store-operated Ca2+ entry [40], where conventional T cell selection is not.

A model of thymic IELp development is shown in Figure 1.

Figure 1. Model of Type A IELp thymic development.

Figure 1.

Self-reactive DP thymocytes can be either clonally deleted or undergo agonist-selection and differentiate to the CD8αα IEL lineage. Costimulation through CD80/86-CD28 is a key factor wherein costimulated cells are deleted, while absence of costimulation can allow clonal diversion of self-reactive thymocytes. Unlike developing conventional T cells, IELp are CCR7 negative and do not migrate to the medulla for further selection processes, but mature in the cortex of the thymus. Maturation of IELp is accompanied by upregulation of CD122 and MHC class-I. Prior to exiting the thymus, IELp become emigration-competent (S1PR1+) and express the mucosal-homing α4β7 integrin. Emigration occurs in an S1P-dependant fashion via perivascular spaces at the corticomedullary junction. Once in the intestine, factors such as intestinal epithelial cell (IEC)-derived IL-15 and food-derived aryl hydrocarbon (AH) ensure the maintenance of CD8αα IEL. Besides Tbet and AH-receptor (AHR), CD8αα IEL express a range of activation and functional markers such as Granzyme B (GzmB), NK receptors of the Ly49 family, and CD160.

A Second Thymic Precursor for CD8αα IEL

While the PD-1+ self-reactive IELp is a prominent and well-studied precursor, recent reports indicate that an alternative and potentially functionally distinct precursor also exists [17,19]. Within the TCRαβ+ DN population, IELp were reported to express either PD-1 [18] or Tbet [30], yet these two populations are mutually exclusive [19]. The PD-1+Tbet (Type A) IELp reside in the thymic cortex, and were reduced in both classical- and nonclassical MHC class-I knockout models (suggesting restriction/crossreactivity to these MHC molecules) and were localized in the thymic cortex. PD-1Tbet+ (Type B) IELp on the other hand were reduced in MHC class-II and nonclassical MHC class-I knockout mice (suggesting restriction/crossreactivity to these MHC molecules), and were mainly found in the medulla. Further, studies with Bim−/− and Nur77GFP mice revealed that Type A cells fall into the classic self-reactive IELp group, while Type B IELp were apparently nonself-reactive [19]. This would fit with observations that some CD8αα IEL recognize microbiota [41], although such cells may also have been derived from CD4+ IEL [42]. Molecular timestamp experiments further indicated that there was no immediate precursor-product relationship between Type A and B IELp. But which of these precursors populate the gut CD8αα IEL pool the most? One study cloned a number of TCRs from CD8αα IEL in the adult gut and expressed them as retrogenics [18]. All of them gave rise to PD-1+ cells in the thymus, leading those authors to conclude that all CD8αα IEL develop from Type A precursors. However, another study compared the TCR repertoires of gut CD8αα IEL to thymic Type A precursors and found a large fraction of specificities that were present in the gut IEL CD8αα IEL but not present in the Type A thymic precursor [21]. Thus, further work on this topic is warranted, as the two populations may be functionally distinct, and there might be disproportionate contributions from each precursor at different ages.

Table 1 outlines some of the distinguishing characteristics of Type A and B IELp.

Table 1. Comparison of two thymic IEL precursors.

Besides self-reactive IELp (Type A), an alternative precursor of CD8αα IEL was defined (Type B). This cell does not show evidence of being self reactive, and expresses distinct cell surface molecules. While the Tbet Type A IELp are localized in the cortex, Tbet+ Type B IELp are found in the medulla. A majority of the thymic emigrant IELp in adult animals are Type A cells.

IELp subset Markers of mature cells Thymic localization TCR-ligand Emigration rate in adults
Type A PD-1highTbetα4β7+ Cortex Self +++
Type B PD-1Tbet+NK1.1+CD103+ Medulla Non-self? +

Emigration and tissue seeding of CD8αα IEL

After selection in the thymus, conventional T cell progenitors upregulate the transcription factor KLF2 and the sphingosine-1 phosphate receptor 1 (S1PR1) and migrate along an S1P gradient through thymic perivascular spaces and into the vasculature [31,4345]. The same emigration mechanism seems to apply to IELp, as the presence of gut CD8αα IEL was dependent on both KLF2 and S1PR1, even though neither of these molecules were expressed by CD8αα IEL in the gut [46]. Instead, KLF2 and S1PR1 are present on thymic IELp, and direct examination of recent thymic emigrant IELp showed their egress to be dependent on these factors [17,19,47]. In fact, thymic S1PR1+ IELp already expressed the mucosal-homing integrin α4β7, suggesting that thymic events program the cell for trafficking to the gut [17,19]. Of interest to the question above, Type A IELp contributed around 90% to the thymic emigrants in adult mice, while Type B only contributed around 10%, despite numeric equivalence in the thymus. Perhaps as a consequence of the slower thymic emigration of Type B compared to Type A IELp, they were found to accumulate in the thymus with age [19].

The ontogeny of CD8αα IEL and their seeding of the intestine is an intriguing subject that warrants further research. It has been shown that CD8αα IEL are established in the small intestine early in life [47,48]. The TCR repertoire of CD8αα IEL is somewhat oligoclonal in nature, being quite variable between each individual [18,49], evoking a scenario where the niche is seeded by a relatively small number of progenitors. The rate of expansion and turnover in early life, and the possibility of temporal differences in the seeding of the gut with qualitatively different CD8αα IEL progenitors is yet unclear. It is conceivable that the onset of solid food uptake, colonization of the gut by microbiota in early life, and/or pathogen exposure could all influence the CD8αα IEL repertoire.

Regarding their maintenance, intestinal epithelial cell-derived IL-15 is known to result in Tbet upregulation, and both are required to maintain CD8αα IEL numbers [30,50]. In addition, aryl hydrocarbon (AH) receptor (AHR)-stimulation through food-derived AH is necessary for the maintenance of CD8αα IEL [9]. Interestingly, components of the microbiota may also activate the AHR, as recently demonstrated in CD4+ IEL for Lactobacillus reuteri-derived products [51]. Another component that deserves further investigation is the β2m-dependent molecules expressed by epithelial cells, such as Qa2, TL, Hfe, or the neonatal Fc receptor, that as mentioned above, may promote local survival and homeostasis.

Future Questions for the Study of CD8αα IEL

In the last decade or so, much effort has been put into better understanding the developmental pathways and functional values of CD8αα IEL. Traditionally, studies of IELp populations heavily rely on adoptive transfer experiments, in which donor populations are injected into congenically distinct T cell deficient (i.e. Rag-1° or Rag-2°) hosts. This approach can rightly be criticized as compromised, as only a small number of IELp can be isolated from thymi, and because they need to be transferred into lymphopenic mice to enable engraftment. A more recent approach is the above mentioned retrogenic mouse model, in which isolated IEL TCR were expressed in Rag° bone marrow cells from a retroviral vector [18,24,25]. While this is an elegant model, studies with retrogenic mice have focused on a limited number of TCRs, usually the most expanded ones. This helps to better understand the big picture but likely does not catch potential minor subpopulations within the heterogeneous IEL pool, which may be different from the dominant populations. To confirm or disprove the existence of various thymic IELp populations, their contribution to total intestinal CD8αα IEL, and for functional studies, retrogenic mice could be utilized by investigating TCR selected from different thymic precursor populations in question. Similarly, receptors from very young versus adult mice could be studied.

In the future, tanscriptomic and epigenetic analysis may be employed to better understand the differentiation, trafficking, and function of CD8αα IEL. Such studies might reveal specific genes that enable the generation of mice allowing for fate mapping studies and selective deficiency models, and allow for more informed comparisons to human IEL populations.

Highlights.

  • Intestinal IEL are a heterogeneous lymphocyte population.

  • In adult animals, the majority of CD8αα IEL precursors are self-reactive thymocytes that undergo agonist selection.

  • Alternative, and likely functionally distinct, precursors exist.

Acknowledgements

KAH received funding from the NIH (R37 AI39560)

Footnotes

Conflict of interest statement: Nothing to declare.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Cheroutre H, Lambolez F, Mucida D: The light and dark sides of intestinal intraepithelial lymphocytes. Nat Rev Immunol 2011, 11:445–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Van Kaer L, Algood HMS, Singh K, Parekh VV, Greer MJ, Piazuelo MB, Weitkamp JH, Matta P, Chaturvedi R, Wilson KT, et al. : CD8alphaalpha(+) innate-type lymphocytes in the intestinal epithelium mediate mucosal immunity. Immunity 2014, 41:451–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Fuchs A, Vermi W, Lee JS, Lonardi S, Gilfillan S, Newberry RD, Cella M, Colonna M: Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-gamma-producing cells. Immunity 2013, 38:769–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.McDonald BD, Jabri B, Bendelac A: Diverse developmental pathways of intestinal intraepithelial lymphocytes. Nat Rev Immunol 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Denning TL, Granger SW, Mucida D, Graddy R, Leclercq G, Zhang W, Honey K, Rasmussen JP, Cheroutre H, Rudensky AY, et al. : Mouse TCRalphabeta+CD8alphaalpha intraepithelial lymphocytes express genes that down-regulate their antigen reactivity and suppress immune responses. J Immunol 2007, 178:4230–4239. [DOI] [PubMed] [Google Scholar]
  • 6.Hoytema van Konijnenburg DP, Reis BS, Pedicord VA, Farache J, Victora GD, Mucida D: Intestinal Epithelial and Intraepithelial T Cell Crosstalk Mediates a Dynamic Response to Infection. Cell 2017, 171:783–794.e713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sujino T, London M, Hoytema van Konijnenburg DP, Rendon T, Buch T, Silva HM, Lafaille JJ, Reis BS, Mucida D: Tissue adaptation of regulatory and intraepithelial CD4(+) T cells controls gut inflammation. Science 2016, 352:1581–1586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shui JW, Larange A, Kim G, Vela JL, Zahner S, Cheroutre H, Kronenberg M: HVEM signalling at mucosal barriers provides host defence against pathogenic bacteria. Nature 2012, 488:222–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, Wilhelm C, Veldhoen M: Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell 2011, 147:629–640. [DOI] [PubMed] [Google Scholar]
  • 10.Cai SF, Fehniger TA, Cao X, Mayer JC, Brune JD, French AR, Ley TJ: Differential expression of granzyme B and C in murine cytotoxic lymphocytes. J Immunol 2009, 182:6287–6297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Leishman AJ, Naidenko OV, Attinger A, Koning F, Lena CJ, Xiong Y, Chang HC, Reinherz E, Kronenberg M, Cheroutre H: T cell responses modulated through interaction between CD8alphaalpha and the nonclassical MHC class I molecule, TL. Science 2001, 294:1936–1939. [DOI] [PubMed] [Google Scholar]
  • 12.Hershberg R, Eghtesady P, Sydora B, Brorson K, Cheroutre H, Modlin R, Kronenberg M: Expression of the thymus leukemia antigen in mouse intestinal epithelium. Proc Natl Acad Sci U S A 1990, 87:9727–9731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Olivares-Villagomez D, Mendez-Fernandez YV, Parekh VV, Lalani S, Vincent TL, Cheroutre H, Van Kaer L: Thymus leukemia antigen controls intraepithelial lymphocyte function and inflammatory bowel disease. Proc Natl Acad Sci U S A 2008, 105:17931–17936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ellmeier W, Sunshine MJ, Maschek R, Littman DR: Combined deletion of CD8 locus cis-regulatory elements affects initiation but not maintenance of CD8 expression. Immunity 2002, 16:623–634. [DOI] [PubMed] [Google Scholar]
  • 15.Rocha B, Vassalli P, Guy-Grand D: The V beta repertoire of mouse gut homodimeric alpha CD8+ intraepithelial T cell receptor alpha/beta + lymphocytes reveals a major extrathymic pathway of T cell differentiation. J Exp Med 1991, 173:483–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Leishman AJ, Gapin L, Capone M, Palmer E, MacDonald HR, Kronenberg M, Cheroutre H: Precursors of functional MHC class I- or class II-restricted CD8alphaalpha(+) T cells are positively selected in the thymus by agonist self-peptides. Immunity 2002, 16:355–364. [DOI] [PubMed] [Google Scholar]
  • 17.Golec DP, Hoeppli RE, Henao Caviedes LM, McCann J, Levings MK, Baldwin TA: Thymic progenitors of TCRalphabeta(+) CD8alphaalpha intestinal intraepithelial lymphocytes require RasGRP1 for development. J Exp Med 2017, 214:2421–2435.* Agonist selection of thymic IELp depends on the Ras activator RasGRP1. Additionaly, this publication implicates the existence of more than one thymic precursor of CD8αα IEL.
  • 18.McDonald BD, Bunker JJ, Ishizuka IE, Jabri B, Bendelac A: Elevated T cell receptor signaling identifies a thymic precursor to the TCRalphabeta(+)CD4(−)CD8beta(−) intraepithelial lymphocyte lineage. Immunity 2014, 41:219–229.** CR from CD8αα IEL in thymocytes showed signs of self-reactivity and induced negative selection. The few cells that escaped clonal deletion became almost exclusively CD8αα IEL, indicating that specific TCR impose an IEL fate.
  • 19.Ruscher R, Kummer RL, Lee YJ, Jameson SC, Hogquist KA: CD8alphaalpha intraepithelial lymphocytes arise from two main thymic precursors. Nat Immunol 2017, 18:771–779.** This study showed that PD-1+ “Type A” IELp are the major precursor to exit the thymus is adult mice. This study also found evidence for an alternative, nonself-reactive “Type B” IELp. These cells differ in their phenotype, MHC-restriction, thymic localization and emigration pattern from self-reactive “Type A” IELp.
  • 20.Stadinski BD, Shekhar K, Gomez-Tourino I, Jung J, Sasaki K, Sewell AK, Peakman M, Chakraborty AK, Huseby ES: Hydrophobic CDR3 residues promote the development of self-reactive T cells. Nat Immunol 2016, 17:946–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wirasinha RC, Singh M, Archer SK, Chan A, Harrison PF, Goodnow CC, Daley SR: alphabeta T-cell receptors with a central CDR3 cysteine are enriched in CD8alphaalpha intraepithelial lymphocytes and their thymic precursors. Immunol Cell Biol 2018, 96:553–561.* The authors show that hydrophobic amino acids such as cysteine in specific positions of the TCR CDR3 were enriched in CD8αα IEL and their precursors.
  • 22.Fujiura Y, Kawaguchi M, Kondo Y, Obana S, Yamamoto H, Nanno M, Ishikawa H: Development of CD8 alpha alpha+ intestinal intraepithelial T cells in beta 2-microglobulin-and/or TAP1-deficient mice. J Immunol 1996, 156:2710–2715. [PubMed] [Google Scholar]
  • 23.Pobezinsky LA, Angelov GS, Tai X, Jeurling S, Van Laethem F, Feigenbaum L, Park JH, Singer A: Clonal deletion and the fate of autoreactive thymocytes that survive negative selection. Nat Immunol 2012, 13:569–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mayans S, Stepniak D, Palida S, Larange A, Dreux J, Arlian B, Shinnakasu R, Kronenberg M, Cheroutre H, Lambolez F: alphabetaT cell receptors expressed by CD4(−)CD8alphabeta(−) intraepithelial T cells drive their fate into a unique lineage with unusual MHC reactivities. Immunity 2014, 41:207–218.** TCR from CD8αα IEL were cloned and their expression forced in thymocytes. Retrogenic cells expressing IEL TCR were shown to be restricted to various MHC types.
  • 25.McDonald BD, Bunker JJ, Erickson SA, Oh-Hora M, Bendelac A: Crossreactive alphabeta T Cell Receptors Are the Predominant Targets of Thymocyte Negative Selection. Immunity 2015, 43:859–869.* This publication showed that crossreactivity to various MHC moleculeswas common among CD8αα IEL and their thymic precursors.
  • 26.Verstichel G, Vermijlen D, Martens L, Goetgeluk G, Brouwer M, Thiault N, Van Caeneghem Y, De Munter S, Weening K, Bonte S, et al. : The checkpoint for agonist selection precedes conventional selection in human thymus. Sci Immunol 2017, 2.* The authors identified a human agonist-selected T cell that has striking similarity to precursors of CD8αα IEL in mice. The findings implicate that CD8αα IEL may also exist in humans and warrants further research on this matter.
  • 27.Poussier P, Julius M: Thymus independent T cell development and selection in the intestinal epithelium. Annu Rev Immunol 1994, 12:521–553. [DOI] [PubMed] [Google Scholar]
  • 28.Lin T, Matsuzaki G, Yoshida H, Kenai H, Omoto K, Umesue M, Singaram C, Nomoto K: Thymus ontogeny and the development of TCR alpha beta intestinal intraepithelial lymphocytes. Cell Immunol 1996, 171:132–139. [DOI] [PubMed] [Google Scholar]
  • 29.Gangadharan D, Lambolez F, Attinger A, Wang-Zhu Y, Sullivan BA, Cheroutre H: Identification of pre- and postselection TCRalphabeta+ intraepithelial lymphocyte precursors in the thymus. Immunity 2006, 25:631–641. [DOI] [PubMed] [Google Scholar]
  • 30.Klose CS, Blatz K, d’Hargues Y, Hernandez PP, Kofoed-Nielsen M, Ripka JF, Ebert K, Arnold SJ, Diefenbach A, Palmer E, et al. : The transcription factor T-bet is induced by IL-15 and thymic agonist selection and controls CD8alphaalpha(+) intraepithelial lymphocyte development. Immunity 2014, 41:230–243.** This report showed dependence of CD8αα IEL on IL-15 and Tbet, and described a Tbet+ thymic precursor of CD8αα IEL.
  • 31.Xing Y, Wang X, Jameson SC, Hogquist KA: Late stages of T cell maturation in the thymus involve NF-kappaB and tonic type I interferon signaling. Nat Immunol 2016, 17:565–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lai YG, Hou MS, Hsu YW, Chang CL, Liou YH, Tsai MH, Lee F, Liao NS: IL-15 does not affect IEL development in the thymus but regulates homeostasis of putative precursors and mature CD8 alpha alpha+ IELs in the intestine. J Immunol 2008, 180:3757–3765. [DOI] [PubMed] [Google Scholar]
  • 33.Ma LJ, Acero LF, Zal T, Schluns KS: Trans-presentation of IL-15 by intestinal epithelial cells drives development of CD8alphaalpha IELs. J Immunol 2009, 183:1044–1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bouillet P, Purton JF, Godfrey DI, Zhang LC, Coultas L, Puthalakath H, Pellegrini M, Cory S, Adams JM, Strasser A: BH3-only Bcl-2 family member Bim is required for apoptosis of autoreactive thymocytes. Nature 2002, 415:922–926. [DOI] [PubMed] [Google Scholar]
  • 35.Stritesky GL, Xing Y, Erickson JR, Kalekar LA, Wang X, Mueller DL, Jameson SC, Hogquist KA: Murine thymic selection quantified using a unique method to capture deleted T cells. Proc Natl Acad Sci U S A 2013, 110:4679–4684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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, 14:377–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, et al. : T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355:1428–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kamphorst AO, Wieland A, Nasti T, Yang S, Zhang R, Barber DL, Konieczny BT, Daugherty CZ, Koenig L, Yu K, et al. : Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 2017, 355:1423–1427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Oh-Hora M, Komatsu N, Pishyareh M, Feske S, Hori S, Taniguchi M, Rao A, Takayanagi H: Agonist-selected T cell development requires strong T cell receptor signaling and store-operated calcium entry. Immunity 2013, 38:881–895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jiang W, Wang X, Zeng B, Liu L, Tardivel A, Wei H, Han J, MacDonald HR, Tschopp J, Tian Z, et al. : Recognition of gut microbiota by NOD2 is essential for the homeostasis of intestinal intraepithelial lymphocytes. J Exp Med 2013, 210:2465–2476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wojciech L, Szurek E, Kuczma M, Cebula A, Elhefnawy WR, Pietrzak M, Rempala G, Ignatowicz L: Non-canonicaly recruited TCRalphabetaCD8alphaalpha IELs recognize microbial antigens. Sci Rep 2018, 8:10848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schwab SR, Pereira JP, Matloubian M, Xu Y, Huang Y, Cyster JG: Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science 2005, 309:1735–1739. [DOI] [PubMed] [Google Scholar]
  • 44.Zachariah MA, Cyster JG: Neural crest-derived pericytes promote egress of mature thymocytes at the corticomedullary junction. Science 2010, 328:1129–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG: Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 2004, 427:355–360. [DOI] [PubMed] [Google Scholar]
  • 46.Odumade OA, Weinreich MA, Jameson SC, Hogquist KA: Kruppel-like factor 2 regulates trafficking and homeostasis of gammadelta T cells. J Immunol 2010, 184:6060–6066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Klose CSN, Hummel JF, Faller L, d’Hargues Y, Ebert K, Tanriver Y: A committed postselection precursor to natural TCRalphabeta(+) intraepithelial lymphocytes. Mucosal Immunol 2018, 11:333–344.* Increasing Tbet expression was shown to correlate with increased IEL fate. In addition, data in this publication suggested that both Tbet+ and Tbet− IELp emigrate from the thymus in an S1P-dependent fashion.
  • 48.Bruce D, Cantorna MT: Intrinsic requirement for the vitamin D receptor in the development of CD8alphaalpha-expressing T cells. J Immunol 2011, 186:2819–2825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Regnault A, Cumano A, Vassalli P, Guy-Grand D, Kourilsky P: Oligoclonal repertoire of the CD8 alpha alpha and the CD8 alpha beta TCR-alpha/beta murine intestinal intraepithelial T lymphocytes: evidence for the random emergence of T cells. J Exp Med 1994, 180:1345–1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Reis BS, Hoytema van Konijnenburg DP, Grivennikov SI, Mucida D: Transcription factor T-bet regulates intraepithelial lymphocyte functional maturation. Immunity 2014, 41:244–256.** This publication highlighted the dependence of CD8αα IEL on Tbet expression.
  • 51.Cervantes-Barragan L, Chai JN, Tianero MD, Di Luccia B, Ahern PP, Merriman J, Cortez VS, Caparon MG, Donia MS, Gilfillan S, et al. : Lactobacillus reuteri induces gut intraepithelial CD4(+)CD8alphaalpha(+) T cells. Science 2017, 357:806–810. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES