首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
Overwhelming evidence suggests that the majority of murine small intestinal intraepithelial lymphocytes (IEL) are extrathymically derived. These IEL include those with T cell receptor (TCR) γδ and someTCR αβ (CD8αα and Thy-1?). In contrast, congenitally athymic nude mice have low numbers of γδ TCR IEL as well as very few αβ TCR IEL, far less than that would be expected if one assumes that γδ TCR IEL and αβ TCR (CD8αα and Thy-1?) IEL in euthymic mice are extrathymically derived. To examine this discrepancy, we followed extrathymic IEL differentiation in IEL of day 3-thymectomized (NTX) mice as another athymic mouse model and found that γδ TCRIEL and extrathymically derived αβ TCR IEL in NTX mice are markedly reduced, almost to the level of nude mice. We further show that it is probably the absence of a thymic stroma that is responsible for the lower amounts of extrathymically derived IEL in nude mice, as the low amounts can be corrected to euthymic levels by syngeneic fetal thymus grafting but not by direct injection of F1 thymocytes. Lastly, unlike TCR/CD3+ extrathymically derived IEL, we noted a large proportion of extrathymic CD3?CD8? and CD3?CD8+ IEL; they were threefold more frequent in nude and NTX than in euthymic mice. This suggests that the thymus influences extrathymically derived IEL in its development from CD3? to CD3+ at the small intestinal epithelium.  相似文献   

2.
We have previously demonstrated that grafting of CBF1(H-2b/d) fetal thymus (FTG) under the kidney capsule of congenitally athymic nude mice of BALB/c background (H-2d) generates a substantial number of T cell receptor (TCR) γδ intestinal intraepithelial lymphocytes (IEL) that were of FTG origin (H-2b+) (see accompanying report). Here we investigated the characteristics of these FTG-derived TCR γδ IEL and compared them to the extrathymically derived TCR γδ IEL found in nude mice. Phenotypically, FTG-derived TCR γδ IEL were similar to their extrathymically derived counterparts in that most were Thy-1 ?, CD5? and CD8αα (homodimer). Vγ and Vδ gene usage in thymus-derived and extrathymically derived TCR γδ IEL were found to be virtually the same. Functionally, FTG-derived TCR γδ IEL were similar to the TCR γδ IEL found in euthymic mice as both were relatively anergic to TCR cross-linking in vitro. However, FTG-derived TCR γδ IEL differed slightly from extrathymically derived TCR γδ IEL, which were completely nonresponsive to the same in vitro stimulation. Overall, these findings support the view that FTG-derived and extrathymically derived TCR γδ IEL are almost indistinguishable. Lastly, we demonstrate that despite their thymic origin, development of FTG-derived TCR γδ IEL partially takes place extrathymically; that is positive selection of FTG-derived Vδ4 IEL occurs extrathymically. In addition, we demonstrate that the CD8 molecule is not necessary for development and homing of FTG-derived TCR γδ IEL. This later finding suggests that the CD8αα molecule develops extrathymically for FTG-derived CD8αα TCR γδ IEL.  相似文献   

3.
Present literature supports the view of an extrathymic origin for the subset of intestinal intraepithelial lymphocytes (IEL) that express the CD4?CD8+ αα phenotype. This subset would include virtually all T cell receptor (TCR) γδ IEL and a portion of TCR αβ IEL. However, these reports do not exclude the possibility that some CD4?CD8+ αα IEL are actually thymically derived. To clarify this issue, we examined the IEL day 3 neonatally thymectomized (NTX) mice. NTX resulted in as much as 80 % reduction in total TCR γδ IEL and in a nearly complete elimination of TCR αβ CD4?CD8+ αα IEL early in ontogeny (3-to 5-week-old mice). The thymus dependency of TCR γδ IEL and TCR αβ CD4?CD8+ IEL was less prominent in older mice (7- to 10-week-old mice), as the total number of these IEL increased in NTX mice, but still remained severalfold less than that in euthymic mice. Furthermore, we demonstrate, by grafting the fetal thymus of CBF1 (H-2b/d) mice under the kidney capsule of congenitally nude athymic mice of BALB/c background (H-2d), that a substantial number of TCR γδ IEL and TCR αβ CD4?CD8+ αα IEL can be thymically derived (H-2b+). In contrast, but consistent with our NTX data, grafting of adult thymi into nude mice generated virtually no TCR γδ IEL and relatively less TCR αβ CD4?CD8+ αα IEL than did the grafting of fetal thymi. These results suggest that the thymus is the major source of TCR γδ and TCR αβ CD4?CD8+ αα IEL early in ontogeny, but that the extrathymic pathway is probably the major source of these IEL later in ontogeny. A reassessment of the theory that most CD4?CD8+ IEL are extrathymically derived is needed.  相似文献   

4.
Intestinal intraepithelial lymphocytes (i-IEL) are phenotypically diverse and consist of both thymically derived and extrathymically derived cells. Extrathymically derived i-IEL are clearly different from thymically derived peripheral T cells in their phenotype and repertoire selection. The major locus of differentiation of extrathymically derived i-IEL appears to be the intestinal epithelium because recombination activating gene (RAG)-1 is expressed in CD3? i-IEL. Extrathymic differentiation however does not imply independence from the thymus as athymic mice have increased numbers of CD3?CD8? and CD3?CD8αα+ i-IEL but decreased numbers of CD3+CD8αα+ i-IEL when compared to euthymic mice. We speculate from these results that thymus-derived cytokine(s)/factor(s) may support differentiation from CD3?CD8αα+ to CD3+CD8αα+ i-IEL in the intestinal epithelium.

I-IEL seem to have some role in immune surveillance because they reside at a site which may represent a first line of defense against pathogenic organisms. This idea is supported by the reports showing in vivo activation of i-IEL under conditions of intestinal infection or tumor-bearing state. In vitro analyses showed cytotoxicity and cytokine production of i-IEL but their true function(s) in vivo is(are) not well known. Clearly more analysis on the in vivo function(s) of i-IEL are needed in order to clarify the true role(s) of i-IEL.  相似文献   

5.
TCRαβ+ CD8α+CD8βintestinal intraepithelial lymphocytes (CD8αα IEL) are gut T cells that maintain barrier surface homeostasis. Most CD8αα IEL are derived from thymic precursors (IELp) through a mechanism referred to as clonal diversion. In this model, self-reactive thymocytes undergo deletion in the presence of CD28 costimulation, but in its absence undergo diversion to the IEL fate. While previous reports showed that IELp were largely β2m dependent, the APC that drive the development of these cells are poorly defined. We found that both CD80 and CD86 restrain IELp development, and conventional DCs play a prominent role. We sought to define a CD80/86 negative, MHCI positive APC that supports the development to the IEL lineage. Chimera studies showed that MHCI needs to be expressed on hematopoietic APC for selection. As thymic hematopoietic APC are heterogeneous in their expression of MHCI and costimulatory molecules, we identified four thymic APC types that were CD80/86neg/low and MHCI+. However, selective depletion of β2m in individual APC suggested functional redundancy. Thus, while hematopoietic APC play a critical role in clonal diversion, no single APC subset is specialized to promote the CD8αα IEL fate.  相似文献   

6.
In conventional mice, the T cell receptor (TCR)αβ+ CD8αα+ and CD8αβ+ subsets of the intestinal intraepithelial lymphocytes (IEL) constitute two subpopulations. Each comprise a few hundred clones expressing apparently random receptor repertoires which are different in individual genetically identical mice (Regnault, A., Cumano, A., Vassalli, P., Guy-Grand, D. and Kourilsky, P., J. Exp. Med. 1994. 180: 1345). We analyzed the repertoire diversity of sorted CD8αα and CD8αβ+ IEL populations from the small intestine of individual germ-free mice that contain ten times less TCRαβ+ T cells than conventional mice. The TCRβ repertoire of the CD8αα and the CD8αβ IEL populations of germ-free adult mice shows the same degree of oligoclonality as that of conventional mice. These results show that the intestinal microflora is not responsible for the repertoire oligoclonality of TCRαβ+ IEL. The presence of the microflora leads to an expansion of clones which arise independently of bacteria. To evaluate the degree of expansion of IEL clones in conventional mice, we went on to measure their clone sizes in vivo by quantitative PCR in the total and in adjacent sections of the small intestine of adult animals. We found that both the CD8αα and the CD8αβ TCRαβ IEL clones have a heterogeneous size pattern, with clones containing from 3 × 103 cells up to 1.2 × 106 cells, the clones being qualitatively and quantitatively different in individual mice. Cells from a given IEL clone are not evenly distributed throughout the length of the small intestine. The observation that the TCRαβ IEL populations comprise a few hundred clones of very heterogeneous size and distribution suggests that they arise from a limited number of precursors, which may be slowly but continuously renewed, and undergo extensive clonal expansion in the epithelium.  相似文献   

7.
We followed αβ T cell receptor (TCR) usage in subsets of gut intraepithelial lymphocytes (IEL) in major histocompatibility complex class I-restricted αβ TCR-transgenic (tg) mice. The proportion of tg αβ TCR+ CD8αβ IEL is reduced compared with CD8+ splenocytes of the same animal, particularly under conventional conditions of maintenance. Further fractionation of CD8αβ IEL according to the expression level of surface CD5 revealed that in conventionally housed animals tg TCR+ CD5? CD8αβ IEL are as frequent as in specific pathogen-free (SPF) mice, whereas tg TCR+ CD5int or, even more pronounced, tg TCR+ CD5hi CD8αβ IEL are greatly diminished when compared with mice kept under SPF conditions. Upon antigen-specific stimulation of CD5? CD8αβ IEL in vitro, CD5 surface expression is up-regulated on a large fraction of cells within 48 h. Up-regulation of CD5 surface expression is further enhanced by the presence of the anti-αIEL monoclonal antibody 2E7. This clearly demonstrates that CD5?, and CD5+ CD8αβ IEL cannot be considered as separate T cell lineages.  相似文献   

8.
《Mucosal immunology》2018,11(5):1281-1289
The location of intraepithelial lymphocytes (IEL) between epithelial cells, their effector memory, cytolytic and inflammatory phenotype positions them to kill infected epithelial cells and protect the intestine against pathogens. Human TCRαβ+CD8αβ+ IEL have the dual capacity to recognize modified self via natural killer (NK) receptors (autoreactivity) as well as foreign antigen via the T cell receptor (TCR), which is accomplished in mouse by two cell subsets, the naturally occurring TCRαβ+CD8αα+ and adaptively induced TCRαβ+CD8αβ+ IEL subsets, respectively. The private/oligoclonal nature of the TCR repertoire of both human and mouse IEL suggests local environmental factors dictate the specificity of IEL responses. The line between sensing of foreign antigens and autoreactivity is blurred for IEL in celiac disease, where recognition of stress ligands by induced activating NK receptors in conjunction with inflammatory signals such as IL-15 can result in low-affinity TCR/non-cognate antigen and NK receptor/stress ligand interactions triggering destruction of intestinal epithelial cells.  相似文献   

9.
The developmental stages and the role of protein tyrosine kinases (PTK) in the maturation of CD3+CD8αα+ intraepithelial lymphocytes (IEL) have not been extensively characterized. However, comparisons of thymic and extrathymic T cell development indicate that these processes involve some distinct signaling and selection events. We used mice deficient in Lck, Fyn, or both Lck and Fyn to analyze the role that these src-family PTK play in IEL development. In contrast to thymocyte development, we found that all IEL subsets develop in mice deficient for either kinase alone. However, lck-/- animals exhibited reduced numbers of TcRαβ+CD8αα+ IEL, indicating that Lck is important in the development of these cells. Mice which lack both Lck and Fyn fail to generate TcRαβ+ IEL, suggesting that signaling through the preTcR, mediated by Lck and, to a lesser extent Fyn, is required for maturation of all TcRαβ+ IEL lineages. Interestingly, a small population of TcRγδ+CD8αα+ cells are apparent in lck-/-fyn-/- animals, demonstrating that TcRαβ+CD8αα+ and TcRγδ+CD8αα+ IEL have distinct PTK requirements for their development or expansion. CD3?-CD8α?CD44+ and CD3?CD8αα+CD16/32+B220+ cells comprise the majority of IEL in both lck-/-fyn-/- and rag-/- mice, while they are poorly represented in wild-type controls. Comparison of the cell surface phenotype of these putative precursor IEL in lck-/-fyn-/- and rag-/- animals suggests that IEL maturation in these animals is arrested at an equivalent developmental stage. Overall, the data presented demonstrate that signals mediated by Lck or Fyn direct TcRαβ+CD8αα+ IEL maturation but are dispensable for the development of TcRγδ+CD8αα+ IEL.  相似文献   

10.
Recent studies in mice and humans have provided evidence for regional specialization of gut intraepithelial lymphocytes (IEL). Here the authors report striking regional variability in the composition of IEL in rat small and large intestine. Two-colour immunofluorescence in situ analysis showed that the distribution of the CD3+ and CD3? IEL subpopulations varied, the proportion of T cells (CD3+) being higher in the ileum than in the jejunum and smallest in the colon. These differences were explained by variable numbers of the T-cell receptor (TCR)α/β+ (both CD8+ and CD4+) but not the TCRγ/δ+ subset. Moreover, the various IEL subpopulations showed distinct intraepithelial distribution patterns with CD4+ and CD8αβ+ T cells situated near the lamina propria, while CD3? IEL were located preferentially towards the adluminal part of the epithelium. Regional phenotypic variation did not depend on intestinal colonization because analogous results were obtained in germ-free rats. Conventionalization nevertheless caused a marked relative increase of small intestinal TCRα/β+ but not TCRγ/δ+ IEL. This increase was more sustained in the jejunum than ileum and eventually reduced the phenotypic IEL differences between the two sites. By contrast, microbial colonization of the colon induced only a transient increase of intraepithelial TCRα/β+ cells with no permanent phenotypic alterations. Both CD3+ and CD3? IEL contained subpopulations that expressed NKR-P1 independent of intestinal colonization. These results demonstrate phenotypic specialization of IEL at different levels of the gut and suggest that the indigenous flora is not essential to this end.  相似文献   

11.
The intestinal lymphoid compartment has a rather stable composition throughout life. However, both during early neonatal development and at high age unique cell populations can be recognized at distinct sites in the intestinal tissue. Directly after birth all intestinal CD3+ cells are found in the lamina propria. At this time the epithelium does not contain T cells. These CD3+ lamina propria lymphocytes co-express both TCRβ and TCRδ chains, probably reflecting the expression of a TCRβδ heterodimer on the cell surface. Cells with this particular phenotype are present in comparable numbers in the lamina propria of both neonatal euthymic and athymic mice, indicating the thymus-independent nature of these cells. During aging the frequency of TCRαβ+ CD8αα+ intestinal T cells increases. These cells are also considered to be thymus-independent. The appearance of high numbers of CD4+ CD8αα+ intestinal T cells in aged mice is even more striking. It is postulated that the neonatal TCRβδ+ cells, and probably also the CD4+ CD8αα+ cells as found in old mice, are intermediates in the extrathymic differentiation pathway of TCRαβ+ CD8αα+ intestinal T cells.  相似文献   

12.
The CD28 antigen has been recently demonstrated to be a costimulatory molecule and is expressed by almost all thymic and peripheral T cell receptor (TcR) αδ+ and γλ+ cells in the mouse system. We show here that expression of CD28 is heterogeneous among murine intestinal intraepithelial lymphocytes (IEL). Whereas some TcR αβ-expressing IEL subsets such as CD4+8? and CD4?+β+ cells express CD28 at the same levels as their phenotypic counterparts in lymph node, other subsets of TcR αβ cells (including CD4?+β? and CD4+8αβ+β cells) as well as TcR γλ+ IEL fail to express CD28. Parallel experiments using aged BALB/c-nu/nu mice indicated that CD28 expression patterns among IEL are quite similar to those of normal BALB/c mice. Furthermore, forward light scatter analysis showed that CD28? cells are considerably larger than CD28+ cells in the gut, although cycling cells were rare in both subsets. Finally CD28? cells in the gut did not proliferate or produce IL-2 upon stimulation by anti-CD3 monoclonal antibodies (mAb) and phorbol 12-myristate 13-acetate, whereas CD28+ cells in the gut and lymph nodes responded to these stimuli. The response of the CD28+ cells was enhanced by anti-CD28 mAb. These results suggest that CD28? IEL (CD4-8α+β? cells, and some CD4++β?cells) may follow a different developmental pathway from that of CD28+ IEL in a thymus-independent environment, and that expression of CD28 correlates with responsiveness of the cells to triggering via the TcR-CD3 complex.  相似文献   

13.
《Mucosal immunology》2021,14(1):68-79
Thymocytes bearing αβ T cell receptors (TCRαβ) with high affinity for self-peptide-MHC complexes undergo negative selection or are diverted to alternate T cell lineages, a process termed agonist selection. Among thymocytes bearing TCRs restricted to MHC class I, agonist selection can lead to the development of precursors that can home to the gut and give rise to CD8αα-expressing intraepithelial lymphocytes (CD8αα IELs). The factors that influence the choice between negative selection versus CD8αα IEL development remain largely unknown. Using a synchronized thymic tissue slice model that supports both negative selection and CD8αα IEL development, we show that the affinity threshold for CD8αα IEL development is higher than for negative selection. We also investigate the impact of peptide presenting cells and cytokines, and the migration patterns associated with these alternative cell fates. Our data highlight the roles of TCR affinity and the thymic microenvironments on T cell fate.  相似文献   

14.
During fetal development, early thymocyte progenitors transiently express low affinity Fc receptors for IgG (FcγR) of both FcγRII and III isoforms. Only the FcγRIII isoform requires association of an FcγRIII (CD16) α subunit with an FcϵRIγ homodimer for surface expression. To address the role of FcγR in ontogeny, we studied thymic development in FcϵRIγ−/− mice. We find that day 14.5 CD4CD8 double-negative (DN) fetal thymocytes of FcϵRIγ−/− mice express mRNA of both FcγRIIb1 and FcγRIII. Surface expression of FcγRII/III is readily detected on these cells. It appears that FcγRIIb1, whose surface expression is FcϵRIγ independent, replaces FcγRIII during thymic development in these animals. Moreover, subsequent development into CD4+CD8+ double-positive and CD4+CD8 and CD4CD8+ single-positive subsets appears normal even in the absence of FcϵRIγ. However, alterations were noted in adult animals among the DN αβ TCR+ thymocytes and peripheral splenic DN T cells as well as CD8αα+ intestinal intraepithelial lymphocytes (iIEL). In contrast to conventional T lymphocytes, which do not express either FcγRIII or FcϵRIγ, DN αβ TCR+ thymocytes and extrathymically derived αβ TCR+ and γδ TCR+ CD8αα+β iIEL express TCR which incorporate FcϵRIγ as one of their subunits. Consistent with this, the TCR levels of these cells are lower than the TCR levels on cells from wild-type C57BL/6 mice. Despite the reduction in the level of surface TCR, the development of these cells was unaltered by the absence of FcϵRIγ. Thus, we observed alterations in adult DN αβ TCR+ thymocytes, splenic DN αβ TCR+ and DN γδ TCR+ large granular lymphocytes (LGL), and αβ TCR+ and γδ TCR+ CD8αα+β iIEL, but no detectable changes in their major fetal thymic developmental pathways. Cultivation of peripheral DN αβ TCR+ and DN γδ TCR+ cells from FcϵRIγ−/− mice with interleukin-2 generates LGL which mediate natural killer activity. Unlike LGL from wild-type C57BL/6 mice, LGL from FcϵRIγ−/− mice lack FcγRIII expression and could not mediate antibody-dependent cellular cytotoxicity through FcγRIII.  相似文献   

15.
Following intraduodenal priming of mice with reovirus, precursor cytotoxic T lymphocytes (pCTL) rapidly appear in intraepithelial lymphocytes (IEL) and Peyer's patches. These cells express CTL activity after secondary in vitro stimulation with reovirus-infected cells. Adoptive transfer of Peyer's patch lymphocytes from normal BALB/c mice into reovirus-infected CB.17 severe combined immunodeficiency mice results in the infection-dependent appearance of large numbers of both CD8+Thy-1+ and CD8?Thy-1+, IEL that express the α/β T cell receptor (TcR). Phenotypic and functional characterization of IEL derived from conventionally reared, reovirus-infected mice also points to extensive similarities in the pCTL derived from Peyer's patches and IEL. As in the Peyer's patches, pCTL are persistent in the IEL compartment for up to 4 weeks after infection. A large percentage of IEL that are recovered from reovirus-primed mice after in vitro culture are CD8+Thy-1+ cells that express α/β TcR. Furthermore, depletion experiments demonstrate that the CD8+Thy-1+ population mediates the virus-specific CTL activity. Using limiting dilution analyses, it was estimated that 7 days after intraduodenal infection the average frequency of virus-specific pCTL was 197/106 CD8+Thy-1+ IEL and 190/106 CD8+Thy-1+ Peyer's patch lymphocytes. Taken together, these observations provide evidence that specific cellular immunity to reovirus in IEL is mediated, at least in part, by conventional cytotoxic T lymphocytes and that these cells are functionally and phenotypically similar to the pCTL derived from the Peyer's patches.  相似文献   

16.
Immunohistochemistry on frozen sections was used to identify CD8αα cells and CD8αβ cells in human intestine. As observed previously, CD8αβ cells predominate (>95%) in tonsil and post-natal intestine. However in human fetal intestine (16–24 weeks gestation), almost half the CD8+ cells in the lamina propria are CD8αα, and many CD8αα cells can be identified in the epithelium. In contrast, in the T cell zones of the Peyer's patches, CD8αβ cells are dominant. The CD8αα cells are virtually all αβ T cell receptor positive. By analogy with the murine system, these CD8αα cells in the fetal gut may be directly derived from the marrow, undergoing thymus-independent differentiation in the gut mucosa.  相似文献   

17.
18.
Clonal selection of T cells mediated through the T cell antigen receptor (TCR) mostly occurs at the CD4+CD8+ double positive thymocyte stage. Immature CD4+CD8+ thymocytes expressing self-reactive TCR are induced to die upon clonotypic engagement of TCR by self antigens. CD3 engagement by antibody of the surface TCR-CD3 complex is known to induce apoptosis of CD4+CD8+ thymocytes, a process that is generally thought to represent antigen-induced negative selection in the thymus. The present study shows that the CD3-induced apoptosis of CD4+CD8+ thymocytes can occur even in TCRα? mutant mice which do not express the TCRαβ/CD3 antigen receptor. Anti-CD3 antibody induces death of CD4+CD8+ thymocytes in TCRα? mice either in cell cultures or upon administration in vivo. Interestingly, most surface CD3 chains expressed on CD4+CD8+ thymocytes from TCRα? mice are not associated with clonotypic TCR chains, including TCRβ. Thus, apoptosis of CD4+CD8+ thymocytes appear to be induced through the CD3 complex even in the absence of clonotypic antigen receptor chains. These results shed light on previously unknown functions of the clonotype-independent CD3 complex expressed on CD4+CD8+ thymocytes, and suggest its function as an apoptotic receptor inducing elimination of developing thymocytes.  相似文献   

19.
20.
《Trends in immunology》2022,43(11):858-860
CD4+ intraepithelial lymphocytes (CD4IEL) are tissue-resident T cells with cytotoxic and regulatory properties; together with peripheral regulatory T cells, they control intestinal inflammation. Recently, Bousbaine and colleagues identified a microbiota-derived conserved antigen that induces CD4IEL differentiation and promotes their regulatory function, attenuating the severity of murine colitis.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号