scholarly journals Enforced Expression of GATA-3 During T Cell Development Inhibits Maturation of CD8 Single-Positive Cells and Induces Thymic Lymphoma in Transgenic Mice

2001 ◽  
Vol 167 (2) ◽  
pp. 715-723 ◽  
Author(s):  
Martijn C. Nawijn ◽  
Rita Ferreira ◽  
Gemma M. Dingjan ◽  
Olev Kahre ◽  
Dubravka Drabek ◽  
...  
1996 ◽  
Vol 183 (4) ◽  
pp. 1707-1718 ◽  
Author(s):  
K F Byth ◽  
L A Conroy ◽  
S Howlett ◽  
A J Smith ◽  
J May ◽  
...  

The CD45 transmembrane glycoprotein has been shown to be a protein phosphotyrosine phosphatase and to be important in signal transduction in T and B lymphocytes. We have employed gene targeting to create a strain of transgenic mice that completely lacks expression of all isoforms of CD45. The spleens from CD45-null mice contain approximately twice the number of B cells and one fifth the number of T cells found in normal controls. The increase in B cell numbers is due to the specific expansion of two B cell subpopulations that express high levels of immunoglobulin (IgM) staining. T cell development is significantly inhibited in CD45-null animals at two distinct stages. The efficiency of the development of CD4-CD8- thymocytes into CD4+ CD8+ thymocytes is reduced by twofold, subsequently the frequency of successful maturation of the double positive population into mature, single positive thymocytes is reduced by a further four- to fivefold. In addition, we demonstrate that CD45-null thymocytes are severely impaired in their apoptotic response to cross-linking signals via T cell receptor (TCR) in fetal thymic organ culture. In contrast, apoptosis can be induced normally in CD45-null thymocytes by non-TCR-mediated signals. Since both positive and negative selection require signals through the TCR complex, these findings suggest that CD45 is an important regulator of signal transduction via the TCR complex at multiple stages of T cell development. CD45 is absolutely required for the transmission of mitogenic signals via IgM and IgD. By contrast, CD45-null B cells proliferate as well as wild-type cells to CD40-mediated signals. The proliferation of B cells in response to CD38 cross-linking is significantly reduced but not abolished by the CD45-null mutation. We conclude that CD45 is not required at any stage during the generation of mature peripheral B cells, however its loss reveals a previously unrecognized role for CD45 in the regulation of certain subpopulations of B cells.


2018 ◽  
Vol 46 (4) ◽  
pp. 441-449
Author(s):  
Sowmya Angusamy ◽  
Tamer Mansour ◽  
Mohammed Abdulmageed ◽  
Rachel Han ◽  
Brian C. Schutte ◽  
...  

Abstract Background: The adaptive immune system of neonates is relatively underdeveloped. The thymus is an essential organ for adaptive T cell development and might be affected during the natural course of oxygen induced lung injury. The effect of prolonged hyperoxia on the thymus, thymocyte and T cell development, and its proliferation has not been studied extensively. Methods: Neonatal mice were exposed to 85% oxygen (hyperoxia) or room air (normoxia) up to 28 days. Flow cytometry using surface markers were used to assay for thymocyte development and proliferation. Results: Mice exposed to prolonged hyperoxia had evidence of lung injury associated alveolar simplification, a significantly lower mean weight, smaller thymic size, lower mean thymocyte count and higher percentage of apoptotic thymocytes. T cells subpopulation in the thymus showed a significant reduction in the count and proliferation of double positive and double negative T cells. There was a significant reduction in the count and proliferation of single positive CD4+ and CD8+ T cells. Conclusions: Prolonged hyperoxia in neonatal mice adversely affected thymic size, thymocyte count and altered the distribution of T cells sub-populations. These results are consistent with the hypothesis that prolonged hyperoxia causes defective development of T cells in the thymus.


1992 ◽  
Vol 2 (2) ◽  
pp. 95-101 ◽  
Author(s):  
Cees de Heer ◽  
Bernard de Geus ◽  
Henk-Jan Schuurma ◽  
Henk Van Loveren ◽  
Jan Rozing

T-cell receptor (TCR)ß-chain usage and expression of the CD3, CD4, and CD8 differentiation antigens were analyzed in 14 spontaneous AKR lymphomas. Lymphoma cells massively infiltrated and/or proliferated in the organs analyzed (thymus, spleen, and mesenteric lymph nodes), giving rise to a loss of organ structure. One lymphoma occurred only in the thymus, and failed to express CD3, CD4, and CD8. All other lymphomas expressed the CD3/TCR complex. With respect to CD4 and CD8 expression, the lymphomas were either double-negative (DN), double-positive (DP), or single-positive (SP). The frequency of DP (CD4+8+) lymphomas was low compared to the frequency of DP thymocytes in a normal AKR thymus. A substantial heterogeneity was seen in the intensity of CD4 and CD8 expression among various lymphomas, which was independent of the level of CD3 expression. Considering TCR Vßgene family usage, 2 out of 14 lymphomas expressed Vß6. Normally, Vß6+thymocytes are deleted from the thymocyte pool at the immature DP stage of T-cell development in AKR mice. These data support the hypothesis that the lymphocytes in the immature DP stage of T-cell development are susceptible to the induction of AKR lymphomagenesis. The presence of Vß6+lymphoma cells indicates that the lymphomagenesis is accompanied by a defective clonal deletion of cells expressing a possible autoreactive TCR.


Blood ◽  
2012 ◽  
Vol 120 (4) ◽  
pp. 789-799 ◽  
Author(s):  
Takeshi Isoda ◽  
Masatoshi Takagi ◽  
Jinhua Piao ◽  
Shun Nakagama ◽  
Masaki Sato ◽  
...  

Immune defect in ataxia telangiectasia patients has been attributed to either the failure of V(D)J recombination or class-switch recombination, and the chromosomal translocation in their lymphoma often involves the TCR gene. The ATM-deficient mouse exhibits fewer CD4 and CD8 single-positive T cells because of a failure to develop from the CD4+CD8+ double-positive phase to the single-positive phase. Although the occurrence of chromosome 14 translocations involving TCR-δ gene in ATM-deficient lymphomas suggests that these are early events in T-cell development, a thorough analysis focusing on early T-cell development has never been performed. Here we demonstrate that ATM-deficient mouse thymocytes are perturbed in passing through the β- or γδ-selection checkpoint, leading in part to the developmental failure of T cells. Detailed karyotype analysis using the in vitro thymocyte development system revealed that RAG-mediated TCR-α/δ locus breaks occur and are left unrepaired during the troublesome β- or γδ-selection checkpoints. By getting through these selection checkpoints, some of the clones with random or nonrandom chromosomal translocations involving TCR-α/δ locus are selected and accumulate. Thus, our study visualized the first step of multistep evolutions toward lymphomagenesis in ATM-deficient thymocytes associated with T-lymphopenia and immunodeficiency.


2002 ◽  
Vol 168 (8) ◽  
pp. 3801-3807 ◽  
Author(s):  
Maryam Yassai ◽  
Kristin Ammon ◽  
Joan Goverman ◽  
Phillipa Marrack ◽  
Yuri Naumov ◽  
...  

Cell ◽  
1990 ◽  
Vol 62 (3) ◽  
pp. 457-467 ◽  
Author(s):  
Robert I. Tepper ◽  
Douglas A. Levinson ◽  
Ben Z. Stanger ◽  
Juanita Campos-Torres ◽  
Abul K. Abbas ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2787-2787
Author(s):  
Mary Philip ◽  
Alexandra R. Zaballa ◽  
Blake T. Hovde ◽  
Janis L. Abkowitz

Abstract Abstract 2787 Heme is essential for nearly every organism and cell. However, free heme can induce free radical formation and cellular damage, therefore cells must carefully regulate heme levels. The feline leukemia virus subgroup C receptor (FLVCR) exports heme from cells. Conditional deletion of Flvcr has been shown to cause progressive anemia in neonatal and adult mice (Science 319:825-8, 2008). Recently, we developed a transplant model in which developing lymphocytes lacked FLVCR while erythroid cells expressed FLVCR, preventing anemia, and found that CD4 and CD8 peripheral T cells were severely decreased while B cell numbers were normal. We further demonstrated that FLVCR-knockout thymocytes were blocked at the CD4CD8 double-positive (DP) stage (Blood [ASH Annual Meeting Abstracts] 114: 913, 2009). We hypothesized that developing T cells lacking FLVCR are arrested at the DP stage because of increased intracellular free heme (IFH). While heme is required for erythroid function, little is known about the role of heme in T cell development. Real-time dynamic quantification of IFH in vivo or from ex vivo tissue is a major challenge in heme biology. We reasoned that by measuring the expression of genes transcriptionally-regulated by heme, we could indirectly assess IFH. Three proteins are key regulators of IFH in non-erythroid cells: aminolevulinic acid synthase-1 (ALAS1) is the rate-limiting enzyme in heme synthesis, FLVCR exports heme, and heme oxygenase-1 (HMOX1) degrades heme. Normal thymic T cell development proceeds from the CD4CD8 double-negative (DN) to the CD4CD8 double-positive (DP) stage, which then go on to either the CD4 single-positive (CD4SP) or CD8 single-positive (CD8SP) stage. We flow-sorted cells from each stage and used multiplex quantitative PCR (qPCR) to determine that all three genes were expressed at higher levels early in normal T cell development during the DN and DP stages and then at lower levels in the CD4SP and CD8SP. Heme binding to the negative regulatory protein BACH1 causes dissociation of BACH1 from the Hmox1 promoter and increased Hmox1 transcription, while expression and stability of Alas1 mRNA is under negative feedback control by heme. Therefore, we predicted that increased IFH in FLVCR-knockout thymocytes would lead to an increase in Hmox1 mRNA and a decrease in Alas1 mRNA levels. We compared expression of heme metabolic genes in FLVCR-knockout and control thymocytes. Flvcr expression was nearly absent in FLVCR-knockout DN and DP cells, however, there was a slight increase in Flvcr expression by the few CD4SP and CD8SP present. To understand this result, we analyzed the extent of genomic Flvcr deletion in FLVCR-knockout thymocytes and peripheral B and T cells by genomic qPCR. DN and DP thymocytes had near complete deletion of Flvcr while CD4SP and CD8SP had slightly less-efficient deletion, likely accounting for the increased Flvcr mRNA levels. Strikingly, Flvcr deletion in the few peripheral T cells present was 50–60% in contrast to peripheral B cells (>90%): only those T cells with incomplete Flvcr deletion survived, further underscoring the absolute requirement for FLVCR in developing T cells. We next examined Hmox1 mRNA expression and found that Hmox1 expression was higher in FLVCR-knockout DP, CD4SP, and CD8SP compared to wild-type FLVCR controls. This supports our hypothesis that FLVCR loss leads to increased IFH during T cell development. Alas1 expression was similar in FLVCR-knockout and control thymocytes, a finding that could be explained because heme regulates ALAS1 activity not only at the transcriptional level but also at the post-transcriptional level. Thus Alas1 expression may not be a good indicator of IFH. In summary, we developed a method to quantify relative free heme levels in developing thymocytes through the measurement of heme metabolic gene expression and found that IFH levels were increased in FLVCR-knockout thymocytes compared to controls. Whether and how excess free heme derails the T cell developmental program, remains to be discovered. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document