scholarly journals Maximum Immunobioactivity of Murine Small Intestinal Intraepithelial Lymphocytes Resides in a Subpopulation of CD43+ T Cells

2004 ◽  
Vol 173 (10) ◽  
pp. 6294-6302 ◽  
Author(s):  
Heuy-Ching Wang ◽  
Dina Montufar-Solis ◽  
Ba-Bie Teng ◽  
John R. Klein
1991 ◽  
Vol 10 (3-4) ◽  
pp. 302-305 ◽  
Author(s):  
Sabine Sarnacki ◽  
Bernadette Bègue ◽  
Anne Jarry ◽  
Nadine Cerf-Bensussan

2003 ◽  
Vol 16 (2) ◽  
pp. 151-156 ◽  
Author(s):  
M.E. Roux ◽  
M.G. Marquez ◽  
S. Olmos ◽  
C.A. Frecha ◽  
A. Florin-Christensen

Compartmentalisation of mucosal immune response seems to be the result mainly of the preferential migration of activated cells back to their inductive sites. The aim of this report was to demonstrate, in a model of secondary immunodeficiency in Wistar rats (severely protein deprived at weaning and refed with casein 20 %; group R21), that the oral administration of Thymomodulin (group: R21TmB) has different effects on gut and BALT (Bronchus-associated lymphoid tissue). Tissue sections (5μ) were studied by immunohistochemistry 1). The oral administration of Thymomodulin restores only in gut Lamina propria (LP) the IgA B and CD4 T cell populations to control levels. The CD8a and CD25 subpopulations do not vary in gut as they return to control levels when refed with 20% casein diet. All the populations mentioned above remained decreased even after receiving Thymomodulin by the oral route. However, the same behaviour was observed for the TCRδ T cells that were decreased and return to normal levels in both mucosae by the effect of the immunomodulator; 2) when studying the iIEL (intestinal intraepithelial lymphocytes) CD8α, CD25 and TCRγδ T cells, that were increased in R21, return to control levels in R21TmB. In BALT intraepithelium CD8α and CD25 T cells remained decreased, while only TCRγδ T cells (increased in R21) return to control values. Conclusions: 1) there exists a compartmentalisation between both mucosae, as T CD4+ and IgA B+ cells are restored by TmB only in gut; 2) only those iIEL involved in inflammation (CD8α+/CD25+ and TCRγδ+/CD25+) are normalised by means of the Thymomodulin 3) however, in BALT, only TCRγδ+ T cells are restored 4) the oral administration of the present immunomodulator may be useful as a therapeutic agent, although the preferential survival in the tissue of initial stimulation is the major factor in the preferential distribution of activated cells.


2000 ◽  
Vol 192 (10) ◽  
pp. 1521-1528 ◽  
Author(s):  
Gobardhan Das ◽  
Dina S. Gould ◽  
Mathew M. Augustine ◽  
Gladis Fragoso ◽  
Edda Scitto ◽  
...  

Murine intestinal intraepithelial lymphocytes (iIELs) are made up of a heterogeneous mix of T cells with unique phenotypes. Whereas CD8+ T cells in peripheral lymphoid organs use CD8α/β and are selected on MHC class Ia molecules, a majority of iIELs use CD8α/α. Here, we report that the presence of CD8α/α TCR-α/β cells in iIELs is independent of classical MHC class I molecules Kb and Db, as illustrated by their presence in Kb/Db double-knockout mice and in mice lacking a nonclassical MHC class I molecule, CD1d. Most strikingly, their presence is decreased by ∼70% in mice lacking transporter associated with antigen processing (TAP). The TAP-dependent nonclassical MHC class I molecule Qa-2 is strongly implicated in the presence of these cells, as inferred from the low numbers of CD8α/α TCR-α/β T cells in mice deficient in Qa-2 genes. Second, a Qa-2–transgenic mouse made in a Qa-2− strain showed an increase in the numbers of CD8α/α cells among its iIELs. Thus, the presence of CD8α/α TCR-α/β cells in iIELs is mainly dependent on the nonclassical MHC class I molecule Qa-2.


1995 ◽  
Vol 182 (1) ◽  
pp. 233-241 ◽  
Author(s):  
D P Hughes ◽  
A Hayday ◽  
J E Craft ◽  
M J Owen ◽  
I N Crispe

Fas-mediated apoptosis is essential for activation-induced cell death of alpha/beta T cells, but it is not clear what role, if any, it plays in regulating other components of the immune system. To study the role of Fas in gamma/delta T cell development, Fas-deficient lpr mice were bred with T cell receptor alpha gene-ablated (TCR-alpha-/-) mice to generate mice deficient in one or both genes. The TCR-alpha-/-, lpr/lpr mice had a nearly 10-fold increase in total lymph node cell (LNC) number compared with Fas-intact TCR-alpha-/- mice, because of expansion of TCR-gamma/delta+ and TCR-beta+ cells. In Fas-intact TCR-alpha-/- mice, approximately one third of the LNCs expressed TCR-gamma/delta. These were evenly divided between the CD4-, CD8-alpha+ and the CD4-, CD8- subsets, and rarely expressed the B220 epitope of CD45. In contrast, in TCR-alpha-/-, lpr/lpr mice, TCR-gamma/delta+ cells comprised half of the LNCs and were primarily CD4-, CD8-, and B220+. Moreover, Fas deficiency in TCR-alpha-/- mice caused a preferential expansion of gamma/delta T cells expressing variable region genes characteristic of intestinal intraepithelial lymphocytes. These results demonstrate a role for Fas in regulating the gamma/delta T cell contribution to peripheral lymph nodes. This mechanism may be most important in limiting the access of activated intestinal intraepithelial lymphocytes to the peripheral lymphoid system.


1998 ◽  
Vol 95 (16) ◽  
pp. 9459-9464 ◽  
Author(s):  
Stephanie T. Page ◽  
Lisa Y. Bogatzki ◽  
Jessica A. Hamerman ◽  
Claire H. Sweenie ◽  
Philip J. Hogarth ◽  
...  

The majority of T cells develop in the thymus and exhibit well characterized phenotypic changes associated with their maturation. Previous analysis of intestinal intraepithelial lymphocytes (IEL) from nude mice and a variety of experimentally manipulated models led to the view that at least a portion of these cells represent a distinct T cell population that matures extrathymically. The IEL that are postulated to mature within the intestine include both T cell receptor (TCR) αβ- and γδ-bearing subpopulations. They can be distinguished from conventional thymically derived T cells in that they express an unusual coreceptor, a CD8α homodimer. In addition, they can utilize the Fc receptor γ-chain in place of the CD3-associated ζ-chain for TCR signaling and their maturation depends on the interleukin 2 receptor β-chain. Moreover, TCRαβ+CD8αα+ IEL are not subject to conventional thymic selection processes. To determine whether CD3−CD8αα+ IEL represent precursors of T cells developing extrathymically, we examined IEL from knockout mice lacking the recombination activating gene-1 (rag-1), CD3ɛ, or both Lck and Fyn, in which thymic T cell development is arrested. CD3−CD8αα+CD16+ IEL from all three mutant strains, as well as from nude mice, included cells that express pre-TCRα transcripts, a marker of T cell commitment. These IEL from lck−/−fyn−/− animals exhibited TCR β-gene rearrangement. However, CD3−CD8αα+CD16+ IEL from ɛ-deficient mice had not undergone Dβ-Jβ joining, despite normal rearrangement at the TCRβ locus in thymocytes from these animals. These results revealed another distinction between thymocytes and IEL, and suggested an unexpectedly early role for CD3ɛ in IEL maturation.


Sign in / Sign up

Export Citation Format

Share Document