scholarly journals Interleukin 7 Transgenic Mice Develop Chronic Colitis with Decreased Interleukin 7 Protein Accumulation in the Colonic Mucosa

1998 ◽  
Vol 187 (3) ◽  
pp. 389-402 ◽  
Author(s):  
Mamoru Watanabe ◽  
Yoshitaka Ueno ◽  
Tomoharu Yajima ◽  
Susumu Okamoto ◽  
Tatsuhiko Hayashi ◽  
...  

We have demonstrated that intestinal epithelial cells produce interleukin 7 (IL-7), and IL-7 serves as a potent regulatory factor for proliferation of intestinal mucosal lymphocytes expressing functional IL-7 receptor. To clarify the mechanism by which locally produced IL-7 regulates the mucosal lymphocytes, we investigated IL-7 transgenic mice. Here we report that transgenic mice expressing murine IL-7 cDNA driver by the SRα promoter developed chronic colitis in concert with the expression of SRα/IL-7 transgene in the colonic mucosa. IL-7 transgenic but not littermate mice developed chronic colitis at 4–12 wk of age, with histopathological similarity to ulcerative colitis in humans. Southern blot hybridization and competitive PCR demonstrated that the expression of IL-7 messenger RNA was increased in the colonic mucosal lymphocytes but not in the colonic epithelial cells. IL-7 protein accumulation was decreased in the goblet cell–depleted colonic epithelium in the transgenic mice. Immunohistochemical and cytokine production analysis showed that lymphoid infiltrates in the lamina propria were dominated by T helper cell type 1 CD4+ T cells. Flow cytometric analysis demonstrated that CD4+ intraepithelial T cells were increased, but T cell receptor γ/δ T cells and CD8α/α cells were not increased in the area of chronic inflammation. Increased IL-7 receptor expression in mucosal lymphocytes was demonstrated in the transgenic mice. These findings suggest that chronic inflammation in the colonic mucosa may be mediated by dysregulation of colonic epithelial cell–derived IL-7, and this murine model of chronic colitis may contribute to the understanding of the pathogenesis of human inflammatory bowel disease.

1993 ◽  
Vol 178 (5) ◽  
pp. 1807-1811 ◽  
Author(s):  
W R Heath ◽  
J F Miller

CD8+ T cells taken directly from mice expressing a Kb-specific T cell receptor (TCR) transgene expressed the transgenic TCR in a bimodal profile as detected by flow cytometric analysis using a clonotype-specific monoclonal antibody. Those cells expressing the lower density of the transgenic TCR expressed the transgenic beta chain and two different alpha chains on their surface. One alpha chain was the product of the alpha transgene, whereas the other was derived by endogenous rearrangement. This report provides the first demonstration that T cells isolated directly from mice may express two different TCR clonotypes on their surface. The potential consequences of this finding for studies using TCR transgenic mice and for the induction of autoimmunity are discussed.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3325-3325
Author(s):  
Alina Ulezko Antonova ◽  
Yitong Wang ◽  
Mojibade Hassan ◽  
Yiwen Li ◽  
Edmund K. Waller

Abstract Introduction: Plasmacytoid dendritic cells (pDC) are known to possess tolerogenic properties in allogeneic bone marrow transplantation (allo-BMT), but the relationship between pDC lineage and their modulation of graft-versus-host disease (GvHD) has not been defined. Recently, we have shown that murine treatment with the pleiotropic cytokine FMS-like Tyrosine Kinase 3 Ligand (Flt3L) increases pDC content in the marrow, and that allo-BMT of Flt3L-treated marrow (FBM) grafts leads to higher overall survival and reduced GvHD in lethally irradiated hosts. Furthermore, FBM pDC have increased potency in preventing GvHD on a cell-by-cell basis (Hassan, EBMT, 2018). Interestingly, FBM pDC expressed CD11b, a myeloid-specific marker that is not expressed on currently defined pDC. Lineage ontogeny of pDC can be tracked based upon exclusive expression of CD31 and Ly6C on myeloid-progenitor derived pDC, while pDC from lymphoid progenitors express RAG1 and Siglec H. We hypothesized that treatment of murine bone marrow donors with Flt3L, which enhances their GvHD-reducing activity, modifies the distribution of lineage-specific precursors of pDC in bone marrow, and that lineage-associated pDC will have distinct biological activity in allo-BMT. Methods and Results: To assess the effect of Flt3L on BM pDC, we treated C57BL/6 mice with Flt3L (CDX-301, 300 ug/kg) on days -1 and -4 relative to bone marrow harvest. Using flow cytometric analysis, we show that FBM pDC over-express myeloid-specific markers CD31, Ly6C and CD11b, but down-regulate the lymphoid-specific marker Siglec H (Figure 1). Moreover, we observed similar phenotypic trends for myeloid-specific markers in pDC from the marrow of RAG1KO mice, confirming the existence of a unique myeloid-derived population of pDC. To determine the effect of pDC linage on their gene expression, we performed Illumina RNA Sequencing on human-derived pDC from Flt3L-mobilized peripheral blood and from human bone marrow. Interestingly, we found that Flt3L-treated pDC overexpress PRSS16, which is known to be exclusively expressed on cortical thymic epithelial cells. Furthermore, using GFP-transgenic mice as a source of donor pDC in B6 -> B10.BR allo-BMT, we show via confocal microscopy that donor pDC selectively home to the recipient thymus (Figure 2). Moreover, flow cytometric analysis revealed that homing of FBM pDC to the thymus is not impaired, in spite of decreased CCR9 expression on FBM pDC in comparison to BM pDC. Because it is currently believed that CCR9 expression on pDC is necessary for their homing to the thymus, these observations denote that FBM pDC use a CCR9-independent homing strategy. Since CD31 expression is upregulated in FBM pDC with enhanced immune-regulatory activity in allo-BMT models, these data suggest that local expression of CD31 by donor pDC in the thymic microenvironment may be relevant to their ability to favorably regulate thymopoiesis and limit the development of alloreactive T cells. Conclusions: Our data poses a link between pDC lineage and control of post-transplant GvHD, possibly via regulation of positive and negative selection of donor-derived T cells in the thymus. Specifically, we identify a CD31+ myeloid-specific population of pDC that emerges in the marrow upon treatment with Flt3L and exhibits a genetic profile that resembles thymic epithelial cells as a candidate for adoptive cell therapy to prevent GvHD. Disclosures Waller: Pharmacyclics: Other: Travel Expenses, EHA, Research Funding; Novartis Pharmaceuticals Corporation: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Kalytera: Consultancy; Celldex: Research Funding; Cambium Medical Technologies: Consultancy, Equity Ownership.


2003 ◽  
Vol 124 (4) ◽  
pp. A35
Author(s):  
Motomi Yamazaki ◽  
Eriko Okada ◽  
Ryuichi Okamoto ◽  
Takanori Kanai ◽  
Hiromichi Ishikawa ◽  
...  

AIDS ◽  
2005 ◽  
Vol 19 (17) ◽  
pp. 1981-1986 ◽  
Author(s):  
François Boutboul ◽  
Denis Puthier ◽  
Victor Appay ◽  
Olivier Pellé ◽  
Hocine Ait-Mohand ◽  
...  

1998 ◽  
Vol 187 (12) ◽  
pp. 2109-2114 ◽  
Author(s):  
H. Robson MacDonald ◽  
Rosemary K. Lees ◽  
Werner Held

Clonally distributed inhibitory receptors negatively regulate natural killer (NK) cell function via specific interactions with allelic forms of major histocompatibility complex (MHC) class I molecules. In the mouse, the Ly-49 family of inhibitory receptors is found not only on NK cells but also on a minor (NK1.1+) T cell subset. Using Ly-49 transgenic mice, we show here that the development of NK1.1+ T cells, in contrast to NK or conventional T cells, is impaired when their Ly-49 receptors engage self-MHC class I molecules. Impaired NK1.1+ T cell development in transgenic mice is associated with a failure to select the appropriate CD1-reactive T cell receptor repertoire. In normal mice, NK1.1+ T cell maturation is accompanied by extinction of Ly-49 receptor expression. Collectively, our data imply that developmentally regulated extinction of inhibitory MHC-specific receptors is required for normal NK1.1+ T cell maturation and selection.


1983 ◽  
Vol 158 (6) ◽  
pp. 1895-1911 ◽  
Author(s):  
D A Cantrell ◽  
K A Smith

T lymphocyte mitosis results from the interaction of interleukin 2 (IL-2) with specific receptors that appear only after appropriate immune stimulation. To assess the potential role of IL-2 receptor levels in determining the rate and magnitude of T cell proliferation, the expression of IL-2 receptors by lectin-stimulated human peripheral blood T cells was examined and correlated with T cell growth. Using biosynthetically radiolabeled IL-2 and anti-Tac, a monoclonal antibody that blocks IL-2 receptor binding, IL-2 receptors were found to accumulate slowly and asynchronously among lectin-stimulated T cells and to precede the onset of DNA synthesis. Moreover, a critical threshold of IL-2 receptor density appeared to be required before the commitment to cell cycle progression, as analyzed quantitatively by tritiated thymidine incorporation and flow cytometric analysis of cellular DNA content. Once maximal IL-2 receptor expression occurred, continued proliferation was IL-2 concentration dependent as assessed using homogenous immunoaffinity-purified IL-2. Upon removal of the activating lectin, IL-2 receptor levels progressively declined, and, in parallel, the rate of proliferation diminished. The decay of IL-2 receptors could not be attributed to IL-2-mediated down-regulation. Instead, renewed IL-2 receptor expression was dependent upon the reintroduction of the initial activating signal. Repetitive exposure to lectin resulted in a more rapid reexpression of maximal IL-2 receptor levels, which was then followed by an accelerated resumption of proliferation. Thus, the extent of T cell proliferation after immune stimulation depends upon the interplay of the IL-2 concentration available and the density of IL-2 receptors expressed, both of which are ultimately determined by antigen/lectin stimulation. The awareness of the transience and the antigen/lectin dependence of IL-2 receptor expression, together with the capacity to monitor T cell cultures for IL-2 receptor levels, should facilitate the initiation and maintenance of cloned, antigen-specific T cells in long-term culture. In addition, these findings suggest that, in vivo, the rapidity of acquisition of maximum IL-2 receptor levels by activated T cells and the duration of IL-2 receptor expression may well direct the magnitude of T cell clonal expansion and resultant immune responses.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 393-393
Author(s):  
Brigitte N. Gomperts ◽  
Marie D. Burdick ◽  
John A. Belperio ◽  
Robert M. Strieter

Abstract Background: Human lung transplants have demonstrated recipient epithelial cells in donor organs, supporting the notion of a circulating epithelial progenitor cell. Resident progenitor epithelial cells in the proximal airway are located in the submucosal gland ducts and basal epithelium and express cytokeratin (CK) 5/14. We have identified circulating progenitor CK5/14 positive epithelial cells in the bone marrow and buffy coat of mice. These cells appear to traffic via the CXCR4/CXCL12 biologic axis. Purpose of study: To further determine additional markers of these circulating progenitor epithelial cells and to disrupt the CXCR4/CXCL12 axis to prevent recruitment of these circulating progenitor cells after airway injury. Methods used: Flow cytometric analysis of cytokeratin 5 (CK5), CXCR4, CD45, CD34, Sca-1, c-kit expressing cells in the buffy-coat and bone marrow of naive mice and CK5-GFP transgenic mice. Subcutaneous heterotopic implantation of dissected female wild type tracheas into male GFP recipient mice. Immunohistochemical and dual immunofluorescence analysis of GFP and CK5 as well as p63 in tracheal transplants. Summary of results: FACS analysis of bone marrow and buffy coat from CK5-GFP transgenic mice and wild type mice revealed that these circulating CK5+ progenitor epithelial cells also express Sca-1,CD34 and c-kit. In the heterotopic tracheal transplant model, wild type tracheal transplantation in CK5-GFP recipient mice demonstrated that the recipient circulating CK5-GFP cells contributed to repair of the airway epithelium. Passive immunization of animals bearing tracheal transplants with specific neutralizing anti-CXCL12 F(Ab)2 antibodies resulted in the unexpected phenotype of the airway epithelium demonstrating squamous metaplasia at day 21 post-transplant.. In contrast, animals bearing tracheal transplants passively treated with control antibodies demonstrated normal pseudostratified epithelium at day 21 post-transplantation. Immunofluorescence for p63 in the tracheal transplants demonstrated that all of the cells of the squamous metaplasia were p63 positive in tracheal transplants from animals that had CXCL12 depleted. Analysis of these tracheal transplants with immunohistochemistry and immunofluorescence for GFP and CK5 demonstrated that the squamous metaplasia was solely derived from resident progenitor epithelial cells. Conclusions: There is a population of CK5+ cells in the bone marrow and circulation of naive mice that also express Sca-1, CD34 and c-kit, which is compatible with a stem cell. These circulating CK5 positive cells contribute to regeneration of the airway epithelium and their recruitment to the progenitor cell niche in the airway appears to be critical for the development of normal pseudostratified columnar epithelium.


2019 ◽  
Vol 3 (s1) ◽  
pp. 4-5
Author(s):  
Eliseo Castillo

OBJECTIVES/SPECIFIC AIMS: The purpose of this research was to investigate how chronic inflammation promotes the generation of proinflammatory intestinal macrophages and if macrophages contribute to intestinal inflammation through Notch activation. METHODS/STUDY POPULATION: We utilized two animal models of chronic colitis, the chronic DSS-induced colitis mouse model and the spontaneous enterocolitis development in IL-10-deficient mice to investigate the role of chronic inflammation in the generation of proinflammatory intestinal macrophages and its influence in notch signaling. Bone marrow-derived monocytes were collected from each group and differentiated into macrophages (BMM) for gene and protein analysis. Ex vivo phenotypical and functional analysis of colonic macrophages was assessed as was the presence of goblet cells and mucosal T cells. In addition, we analyzed the development of goblet cell differentiation in colonoids in a co-culture system with proinflammatory macrophages. RESULTS/ANTICIPATED RESULTS: Our chronic inflammation models revealed an increase in proinflammatory macrophages present in the lamina propria and that these cells expressed significantly higher levels of notch ligand, Jagged1. Jagged 1 has been shown to enhance TH1 differentiation and T cells isolated from the mucosa of both chronic colitis models display strong TH1 skewing compared to controls. Chronic inflammation also contributes to intestinal barrier defects, enhanced permeability and bacterial translocation. We believe this enhanced intestinal permeability and subsequent bacterial translocation promote Jagged1 expression in intestinal macrophages. To support this concept, we show TLR stimulation induces the upregulation of Jagged1 in BMM. Additionally, the generation of BMM from our chronic DSS-induced colitis mice or age matched controls, revealed BMM derived from a host of chronic inflammation were skewed to a proinflammatory state prior to stimulation showing increased gene expression of several proinflammatory molecules including IL-1α, IL-1β, IL-12 and TNF-α. This would suggest monocytes migrating to the intestinal mucosa have more potential to become proinflammatory instead of traditional anti-inflammatory macrophages. Furthermore, proinflammatory notch ligand-positive macrophages co-cultured with colonoids, derived from unperturbed mice, significantly decreased the number of mucus producing goblet cells. In support of this observation, notch activation in intestinal stem cells promote absorptive (i.e. colonocytes) cell differentiation and prevents secretory cell (i.e. goblet cells) differentiation. DISCUSSION/SIGNIFICANCE OF IMPACT: Taken together, our results strongly suggest chronic inflammation modulates macrophages role in maintaining intestinal homeostasis through possible notch activation in both T cells and the intestinal epithelial barrier.


2006 ◽  
Vol 74 (4) ◽  
pp. 2495-2497 ◽  
Author(s):  
Alexandre Morrot ◽  
Ian A. Cockburn ◽  
Michael Overstreet ◽  
Dolores Rodríguez ◽  
Fidel Zavala

ABSTRACT CD8+ T cells induced by Plasmodium yoelii sporozoites develop into protective memory cells without undergoing changes in interleukin-7 receptor α (IL-7Rα) expression, differing from the development of memory CD8+ T cells against viruses, which is associated with enhanced IL-7Rα expression. This suggests a microbe-dependent diversity in the signals determining the development of memory populations.


Sign in / Sign up

Export Citation Format

Share Document