A Rare Population of Stem Cell Memory T Cells is an Apex of the Hierarchy of Adult T-Cell Leukemia

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3001-3001
Author(s):  
Yuya Nagai ◽  
Masahiro Kawahara ◽  
Masakatsu Hishizawa ◽  
Noriko Sugino ◽  
Yayoi Shimazu ◽  
...  

Abstract Adult T-cell leukemia (ATL) is a mature T cell neoplasm causatively associated with HTLV-1. It takes a long period, around 50 years, to develop ATL after the transmission of HTLV-1 in infancy through breastfeeding, suggesting the pool for the generation of ATL clone may exist somewhere. Recent reports showed human CD34+ bone marrow cells infected by HTLV-1 ex vivo can generate CD4+ T-cell lymphomas in immunodeficiency mice or that CD117+ early hematopoietic progenitors can develop lymphoma in transgenic mice model of Tax which is an oncogenic protein of HTLV-1. These results suggest the model of leukemia initiating cells, that leukemia is organized hierarchically and sustained by a small subset of stem/progenitors at the apex, might be acceptable in ATL. A more recent report demonstrated that a novel compartment of normal T cells, termed stem cell memory T cells (TSCM), possesses stem cell-like capacity including relatively enhanced self-renewal capacity and multipotent ability to generate all memory and effector T cell subsets. This concept prompts us to hypothesize that TSCM subset might contribute to the development of ATL. First, to investigate whether hematopoietic stem/progenitor cells in primary samples of ATL patients are infected with HTLV-1, we sorted lineage(CD3, CD4, CD8, CD11b, CD14, CD19, CD20, CD56, CD235)-negative CD34+ cells from bone marrow samples of ATL patients by FACS AriaII, purified genomic DNA, and then performed PCR of pX region which is a virus specific gene. No amplification of pX was detected in all samples we studied. Next, to focus on TSCM population, we established the eight-color staining method with lineage of NK/Monocyte/DC (CD11b, CD14 and CD56), CD4, CD45RA, CD45RO, CCR7, CD95, CD122 and CD25. We successfully separated CD4+ T cells from ATL patients into four subsets; naïve T (TN: CD4+CD45RA+CD45RO-CCR7+CD95-CD122-), stem cell memory T (TSCM: CD4+CD45RA+CD45RO-CCR7+CD95+CD122dim), central memory T (TCM: CD4+CD45RA-CD45RO+CCR7+), and effector memory T (TEM: CD4+CD45RA-CD45RO+CCR7-). Although ATL cells are generally CD45RO positive, so that TCM and TEM were extremely dominant, CD45RA positive populations did exist in all ATL samples we studied. To assess the frequency of infected cells, we performed quantitative PCR with the sensitivity of 5 copies detectable at the minimum and calculated by dividing copy number of pX in HTLV-1 provirus with that of RNase P in human genome in genomic DNA from 1,000 cells of each subset. Almost all cells in TCM and TEM and more than 10% of cells in TSCM were infected in most of cases while none of infected cells were detected in TN in our experiment design. Since ATL is caused by monoclonal expansion from plenty of HTLV-1 infected clones, we focused on the existence of ATL original clone in each subset. We determined the ATL original clone by inverse PCR and the clone-specific provirus integration site by sequencing of the inverse PCR product in each case, and then constructed PCR primers being capable of detecting each ATL original clone by regular PCR. Interestingly we found each ATL clone was detected in TSCM, TCM, and TEM but not in TN subset in all studied cases, indicating TSCM could be an origin of ATL cells. To investigate whether there is a hierarchy in ATL cells, we next performed in vitro culture with IL-7 and examined the status of CD45RA and CCR7 after 2 weeks. Intriguingly TSCM generated TCM and TEM while TCM and TEM did not generate TSCM, suggesting TSCM is an apex in HTLV-1 infected T cells. To investigate tumorigenicity of TSCM, we adopted xenogeneic transplantation assay. When we inoculated 3x104 cells of each subset from one ATL patient into immune deficient mice (NOG mice) intraperitoneally, we observed high number of cells in peripheral blood and spleen of recipient mice transplanted with TSCM compared to TCM and TEM. Phenotypically TCM and TEM were generated in mice transplanted with TSCM as well as TCM and TEM while TSCM was only generated from TSCM but not TCM and TEM. Furthermore we analyzed the clonality of engrafted cells by ATL clone-specific PCR and inverse PCR, and found that the patient original clone becomes dominant in recipient mice inoculated with TSCM but not in those with TCM and TEM. Taken together, our findings provide new insights that TSCM could be an apex of the hierarchy of ATL and play an important role in development and maintenance of ATL clone. Disclosures: No relevant conflicts of interest to declare.

2018 ◽  
Vol 7 (5) ◽  
pp. e1426423 ◽  
Author(s):  
Ricardo Khouri ◽  
Gilvanéia Silva-Santos ◽  
Tim Dierckx ◽  
Soraya Maria Menezes ◽  
Daniele Decanine ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4144-4144
Author(s):  
Yamin Tian ◽  
Seiichiro Kobayashi ◽  
Nobuhiro Ohno ◽  
Masamichi Isobe ◽  
Mayuko Tsuda ◽  
...  

Abstract Abstract 4144 [Background] Adult T-cell leukemia (ATL) is a malignant disorder caused by human T-cell leukemia virus type I (HTLV-I). Morphological discrimination of leukemic cells from non-leukemic T cells is often difficult in ATL since ATL cells reveal morphological diversity except for typical “flower cells”. Although a study using CD3 gating in flow cytometry reported that ATL cells were distinguishable as a CD3low population from normal lymphocytes, these cells were not well characterised as ATL cells. Considering that defective expression of CD7 as well as CD3 is common in ATL cells, we applied multi-color flow cytometry to detect a putative leukemia-specific cell population in the peripheral blood from ATL patients. [Methods and Results] (1) In flow cytometry, after dead-cell and monocyte removal, CD4+ T lymphocytes were gated on the CD3 versus CD4 plot. Based on cell density and fluorescence intensity of CD3 and CD7 in this population, we designated three subpopulations on this plot: CD3highCD7high, CD3dimCD7dim and CD3dimCD7low(Results of a representative ATL and a control sample are shown in Figure). The proportion of the CD3dim/CD7low subpopulation was significantly higher in acute-type ATL CD4+ lymphocytes than in normal controls(Figure). (2) To extensively characterise this subpopulation, we next estimated the HTLV-I proviral load by quantitative real-time PCR after FACS sorting based on this CD3 versus CD7 plot. In all patient samples, HTLV-I proviral integration was detected in all subpopulations. However, the proviral load was significantly higher in the CD3dim/CD7low subpopulation compared to the CD3high/CD7high subpopulation. Almost all of the cells in the CD3dim/CD7low subpopulation were HTLV-I infected. (3) We next examined CCR4 and CD25 expression in each subpopulation. Both CCR4 and CD25 expression levels were maintained at very low and similar levels throughout all subpopulations in normal control cells and in the CD3high/CD7high subpopulation of patients with ATL as well. In contrast, CCR4 expression was significantly up-regulated in CD3dim/CD7low subpopulation of patients with ATL compared to the CD3high/CD7high subpopulation (MFI: 36.5±17.2 vs. 3.8±1.1). The expression of CD25 was also up-regulated in the subpopulation (MFI: 7.8±8.0 vs. 2.7±1.6). (4) Monoclonal expansion of HTLV-I-infected cells in the CD3dim/CD7low subpopulation was indicated by the genomic integration site analysis using a long inverse polymerase chain reaction (PCR) method. (5) We reviewed the glass-slide specimens of FACS-sorted samples to evaluate the morphology of each subpopulation on the CD3 versus CD7 plot. Atypical lymphocytes with morphology such as a notch in the nucleus were observed in all subpopulations. The majority of sorted cells from CD3dim/CD7low subpopulation showed “flower cell”-like morphology. (6) We also detected a small CD3dim/CD7dim subpopulation other than the CD3dim/CD7low and CD3high/CD7high subpopulations in all patients with acute-type ATL who were analysed(Figure). This subpopulation contained the same clone as the CD3dim/CD7low subpopulation, although a phenotypical difference existed between these subpopulations. [Conclusion] (1) Above findings indicate that leukemic T cells are specifically enriched in a unique CD3dim/CD7low subpopulation of CD4+ T cells in acute-type ATL. This multi-color FACS system may be useful for precisely monitoring disease during chemotherapy, detecting minimal residual disease and analysing ATL cells. (2) Previous reports have revealed that HTLV-I-infected cells transform through multi-step oncogenesis. Detailed analysis of these three subpopulations (CD3high/CD7high, CD3dim/CD7dim and CD3dim/CD7low) may give some insight into oncogenesis of HTLV-I-infected cells. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 ◽  
Author(s):  
Thomas J. Weitering ◽  
Janine E. Melsen ◽  
Monique M. van Ostaijen-ten Dam ◽  
Corry M. R. Weemaes ◽  
Marco W. Schilham ◽  
...  

Ataxia Telangiectasia (AT) is a rare inherited disorder characterized by progressive cerebellar ataxia, chromosomal instability, cancer susceptibility and immunodeficiency. AT is caused by mutations in the ATM gene, which is involved in multiple processes linked to DNA double strand break repair. Immunologically, ATM mutations lead to hampered V(D)J recombination and consequently reduced numbers of naive B and T cells. In addition, class switch recombination is disturbed resulting in antibody deficiency causing common, mostly sinopulmonary, bacterial infections. Yet, AT patients in general have no clinical T cell associated infections and numbers of memory T cells are usually normal. In this study we investigated the naive and memory T cell compartment in five patients with classical AT and compared them with five healthy controls using a 24-color antibody panel and spectral flow cytometry. Multidimensional analysis of CD4 and CD8 TCRαβ+ cells revealed that early naive T cell populations, i.e. CD4+CD31+ recent thymic emigrants and CD8+CCR7++CD45RA++ T cells, were strongly reduced in AT patients. However, we identified normal numbers of stem cell memory T cells expressing CD95, which are antigen-experienced T cells that can persist for decades because of their self-renewal capacity. We hypothesize that the presence of stem cell memory T cells explains why AT patients have an intact memory T cell compartment. In line with this novel finding, memory T cells of AT patients were normal in number and expressed chemokine receptors, activating and inhibitory receptors in comparable percentages as controls. Comparing memory T cell phenotypes by Boolean gating revealed similar diversity indices in AT compared to controls. We conclude that AT patients have a fully developed memory T cell compartment despite strongly reduced naive T cells. This could be explained by the presence of normal numbers of stem cell memory T cells in the naive T cell compartment, which support the maintenance of the memory T cells. The identification of stem cell memory T cells via our spectral flow cytometric approach is highly relevant for better understanding of T cell immunity in AT. Moreover, it provides possibilities for further research on this recently identified T cell population in other inborn errors of immunity.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2516-2516
Author(s):  
Haruka Kinosada ◽  
Jun-ichirou Yasunaga ◽  
Kazuya Shimura ◽  
Masao Matsuoka

Abstract Introduction Adult T-cell leukemia (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1). It has been reported that co-inhibitory receptors such as programmed cell death 1 (PD-1) and T cell immunoglobulin and ITIM domain (TIGIT) are highly expressed on ATL cells and HTLV-1 infected cells. However, ATL cells and HTLV-1 infected cells proliferate regardless of their high expression. Although it has been known that HTLV-1 bZIP factor (HBZ), which is constitutively expressed in ATL cells and HTLV-1 infected cells, promotes T cell proliferation, the detailed mechanisms remain unclear. In this study, we found that HBZ promotes T-cell proliferation by interfering the suppressive function of co-inhibitory receptors. Methods We analyzed T-cell proliferation of HBZ transgenic (HBZ-Tg) mice that specifically express HBZ in CD4+ T cells, and expression of co-inhibitory and co-stimulatory molecules on ATL cells and CD4+ T cells of HBZ-Tg mice. Furthermore, the function of TIGIT and PD-1 was studied using HBZ-transduced murine CD4+T cells. The co-localization of SHP-2 and PD-1 in the presence of HBZ was analyzed by immunoprecipitation and confocal microscope. The immunoprecipitation and confocal microscope were also used to analyze interaction between HBZ and THEMIS and HBZ localization in the presence of THEMIS. Results Although HBZ promotes T-cell proliferation, we found that some co-inhibitory receptors, TIGIT and PD-1, were highly expressed on CD4+ T cells of HBZ-Tg mice and ATL cells. As mechanisms, HBZ upregulated transcriptions of these genes. Based on these observations, we hypothesized that HBZ impairs suppressive functions of TIGIT and PD-1 while it increases expression of these co-inhibitory receptors. To address this question, we analyzed the suppressive activity of TIGIT and PD-1 in the presence of HBZ. We transduced HBZ by the retrovirus vector into primary murine T cells and evaluated the proliferation after stimulated TIGIT or PD-1 with anti-CD3 and its ligand. As a result, TIGIT and PD-1 did not inhibit T-cell proliferation in the presence of HBZ, indicating that HBZ impairs the suppressive function of TIGIT and PD-1. Both TIGIT and PD-1 possess SHP-2, a tyrosine phosphatase, binding domains in its cytoplasmic tail, ITIM or ITSM motif. Therefore, we next studied whether HBZ influences the interaction between PD-1 and SHP-2. Tyrosine phosphorylation of PD-1 was induced with pervanadate and then SHP-2 recruitment and PD-1/SHP-2 co-localization were investigated. HBZ inhibited recruitment of SHP-2 to the ITSM motif of PD-1. Indeed, phosphorylation of SHP-2 was decreased in CD4+T cells of HBZ-Tg mice and HBZ-transduced murine primary T cells. Furthermore, function of SHP-2 to dephosphorylate ZAP-70 and CD3-zeta was suppressed in the presence of HBZ. These data showed that HBZ inhibited recruitment of SHP-2 to ITSM motif of PD-1 and suppressed its inhibitory function. Next, we examined how HBZ inhibits the interaction between PD-1 and SHP-2. HBZ did this by interacting with THEMIS, which forms a complex with Grb2 and SHP-2. Moreover, HBZ hindered the interaction between THEMIS and Grb2. In general, THEMIS is localized in the cytoplasm, whereas it has been reported that HBZ is localized in the nucleus. When we expressed THEMIS or HBZ, THEMIS existed in the cytoplasm (50 of 50 cells: 100%) whereas HBZ was mainly localized in the nucleus (67 of 74 cells: 90.5%). Interestingly, when both proteins were expressed in the same cells, HBZ changed its localization to cytoplasm (28 of 79 cells: 35.4%) and co-localized with THEMIS. These findings suggest that THEMIS changes the localization of HBZ from nucleus to cytoplasm. Thus, HBZ functions not only in the nucleus but also in the cytoplasm by interacting with host factors. Since THEMIS is expressed only in T-lineage cells, inhibition of the suppressive effects of co-inhibitory receptors by HBZ accounts for how HTLV-1 induced proliferation only T cells in vivo. Conclusions Our findings demonstrated that HBZ promotes T-cell proliferation upon TCR stimulation by impairing the suppressive signal of co-inhibitory receptors. This study presents the first evidence of mechanisms how HBZ attenuates the inhibitory signals and promotes T-cell proliferation. Disclosures No relevant conflicts of interest to declare.


Retrovirology ◽  
2014 ◽  
Vol 11 (1) ◽  
Author(s):  
Kieran Cashin ◽  
Geza Paukovics ◽  
Martin R Jakobsen ◽  
Lars Østergaard ◽  
Melissa J Churchill ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document