scholarly journals HL-T, a new cell line derived from HL-60 promyelocytic leukemia cell cultures expressing terminal transferase and secreting suppressor activity

Blood ◽  
1987 ◽  
Vol 70 (4) ◽  
pp. 1151-1160 ◽  
Author(s):  
E Paietta ◽  
RJ Stockert ◽  
T Calvelli ◽  
P Papenhausen ◽  
SV Seremetis ◽  
...  

Abstract A cell line with immature blast cell morphology was isolated from HL-60 promyelocytic leukemia cell cultures and designated HL-T. This new cell type is biphenotypic, expressing terminal transferase (TdT) together with myelomonocytoid immunologic features. TdT enzymatic activity, undetectable in HL-60, was determined to be 140 to 180 units/10(8) HL-T cells by the dGTP-assay, approximately 20% of the activity found in lymphoblastoid cell lines. HL-T predominantly synthesize the known 58- kDa TdT-protein plus a minor 54/56-kDa doublet. The 58-kDa steady state form is nonglycosylated and is phosphorylated. Precursor antigens S3.13 and MY-10, absent on HL-60, are expressed by HL-T; however, the cells are negative for HLA-Dr. Southern blot analysis by hybridization with immunoglobulin heavy chain (JH) and T cell-receptor chain gene (T beta) probes shows JH to be in the germ-line configuration in both cell lines and the T beta gene to be in germ-line in HL-60 but to be rearranged in HL-T. Truncation of the gene encoding the granulocyte-macrophage-colony- stimulating factor (GM-CSF), as found in HL-60, is not observed in HL- T. HL-T are resistant to differentiation-induction by retinoic acid and 1,25-dihydroxyvitamin D3. Cytogenetically HL-T share with HL-60 a deletion of the short arm of chromosome 9 at breakpoint p13, an aberration frequently found in patients with T cell leukemia. In addition, HL-T display t(8;9)(p11;p24) and trisomy 20. Tetraploidy is observed in 80% of HL-T metaphases with aberrations identical to those in the diploid karyotype. Like HL-60, the new line shows some surface- antigenic-T cell characteristics. Despite an antigenic pattern most consistent with that of helper-inducer T cells (T4+, D44+/-, 4B4+, 2H4- , TQ1+/-), HL-T cells and their conditioned culture medium suppress antigen, mitogen, and mixed-leukocyte-culture-mediated lymphocyte proliferation.

Blood ◽  
1987 ◽  
Vol 70 (4) ◽  
pp. 1151-1160 ◽  
Author(s):  
E Paietta ◽  
RJ Stockert ◽  
T Calvelli ◽  
P Papenhausen ◽  
SV Seremetis ◽  
...  

A cell line with immature blast cell morphology was isolated from HL-60 promyelocytic leukemia cell cultures and designated HL-T. This new cell type is biphenotypic, expressing terminal transferase (TdT) together with myelomonocytoid immunologic features. TdT enzymatic activity, undetectable in HL-60, was determined to be 140 to 180 units/10(8) HL-T cells by the dGTP-assay, approximately 20% of the activity found in lymphoblastoid cell lines. HL-T predominantly synthesize the known 58- kDa TdT-protein plus a minor 54/56-kDa doublet. The 58-kDa steady state form is nonglycosylated and is phosphorylated. Precursor antigens S3.13 and MY-10, absent on HL-60, are expressed by HL-T; however, the cells are negative for HLA-Dr. Southern blot analysis by hybridization with immunoglobulin heavy chain (JH) and T cell-receptor chain gene (T beta) probes shows JH to be in the germ-line configuration in both cell lines and the T beta gene to be in germ-line in HL-60 but to be rearranged in HL-T. Truncation of the gene encoding the granulocyte-macrophage-colony- stimulating factor (GM-CSF), as found in HL-60, is not observed in HL- T. HL-T are resistant to differentiation-induction by retinoic acid and 1,25-dihydroxyvitamin D3. Cytogenetically HL-T share with HL-60 a deletion of the short arm of chromosome 9 at breakpoint p13, an aberration frequently found in patients with T cell leukemia. In addition, HL-T display t(8;9)(p11;p24) and trisomy 20. Tetraploidy is observed in 80% of HL-T metaphases with aberrations identical to those in the diploid karyotype. Like HL-60, the new line shows some surface- antigenic-T cell characteristics. Despite an antigenic pattern most consistent with that of helper-inducer T cells (T4+, D44+/-, 4B4+, 2H4- , TQ1+/-), HL-T cells and their conditioned culture medium suppress antigen, mitogen, and mixed-leukocyte-culture-mediated lymphocyte proliferation.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2666-2666
Author(s):  
P.K. Epling-Burnette ◽  
Jeffrey S. Painter ◽  
Fanqi Bai ◽  
Subra Mohaptra ◽  
Thomas P. Loughran

Abstract Low-dose methotrexate (MTX) is used as an immunosuppressive agent for the treatment of rheumatoid arthritis (RA), Large Granular Lymphocyte (LGL) leukemia, Cutaneous T Cell Lymphoma (CTCL), autoimmune diseases, and prevention of GvHD during bone marrow transplants. The mechanism for immunosuppression is not clearly understood but most data suggests that apoptosis of activated lymphocytes plays a critical role. In this study, we wanted to define the MTX-sensitive population and to determine the apoptotic pathway activated by MTX. Using a clinically relevant dosage range (8 nM- 1 μM), MTX-mediated apoptosis was first examined in a T lymphoblastic leukemia cell line (CEM). The apoptotic pathway induced by MTX included phosphotidylinositol externalization and caspase-3 activation along with a slight increase in mitochondrial membrane depolarization. We next examined a series of tumor cell lines and normal cells for evidence of MTX-induced apoptosis. Using the same clinically relevant dosage range, we found that MTX-induced apoptosis was primarily observed in the four T cell leukemia cell lines including CEM, Jurkat, MT-2, and HUT78 and in normal PBMCs activated with mitogens and IL-2. Less MTX-induced apoptosis was observed in two myeloid leukemia cell lines including HL-60 and K562 and in a B cell leukemia cell line Raji, and the multiple myeloma cell line 8226. Unactivated PBMCs were resistant to MTX-mediated apoptosis. T cells that are clonally expanded in patients with T-LGL leukemia have a CD8+ cytotoxic phenotype, whereas other diseases that are treated with low-dose MTX, such as CTCL and RA, are characterized by the expansion of CD4+ T cells. We found that both freshly sorted CD4+ and CD8+ cells were MTX resistant. In contrast, PHA plus IL-2 treatment induced MTX sensitivity in T cell with both immunophenotypes. We also examined clinical samples from patients with LGL leukemia. We found that freshly isolated PBMCs from T-LGL leukemia patients were resistant to MTX. Clonal cells from the peripheral blood of LGL leukemia patients are in G0/G1 phase of the cell cycle. Interestingly, we found that PHA plus IL-2 treatment induced the cells to enter S-phase and to become MTX sensitive. These results suggest that only fully activated, proliferating T cells from patients with LGL leukemia undergo apoptosis in response to low-dose MTX. Because there was only minor depolarization of mitochondria after MTX treatment in both CEM cells and normal activated PBMCs, we wanted to examine upstream apoptotic events after MTX treatment. We found that caspase-8 cleavage and enzymatic activity was induced by MTX in both CD95 Type I (HUT78) and Type II (CEM and Jurkat) cells but that there was a differential requirement for caspase-8 activity for apoptosis. We found that caspase-8 activation was independent of the Fas receptor as shown by immunoprecipitation experiments and MTX apoptotic assays in the JM3A5 Fas-receptor mutant Jurkat cell line. Using a Jurkat cell line with a homozygous deletion of the FADD gene, we found that caspase-8 activation, caspase-3 activation, and apoptosis in response to MTX were dependent on the adaptor protein FADD. These findings have important implications for understanding the mechanism of MTX for immunosuppressive therapy.


1994 ◽  
Vol 14 (11) ◽  
pp. 7604-7610
Author(s):  
H M Pomykala ◽  
S K Bohlander ◽  
P L Broeker ◽  
O I Olopade ◽  
M O Díaz

Interstitial deletions of the short arm of chromosome 9 are associated with glioma, acute lymphoblastic leukemia, melanoma, mesothelioma, lung cancer, and bladder cancer. The distal breakpoints of the deletions (in relation to the centromere) in 14 glioma and leukemia cell lines have been mapped within the 400 kb IFN gene cluster located at band 9p21. To obtain information about the mechanism of these deletions, we have isolated and analyzed the nucleotide sequences at the breakpoint junctions in two glioma-derived cell lines. The A1235 cell line has a complex rearrangement of chromosome 9, including a deletion and an inversion that results in two breakpoint junctions. Both breakpoints of the distal inversion junction occurred within AT-rich regions. In the A172 cell line, a tandem heptamer repeat was found on either side of the deletion breakpoint junction. The distal breakpoint occurred 5' of IFNA2; the 256 bp sequenced from the proximal side of the breakpoint revealed 95% homology to long interspersed nuclear elements. One- and two-base-pair overlaps were observed at these junctions. The possible role of sequence overlaps, and repetitive sequences, in the rearrangement is discussed.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 28-29
Author(s):  
Jie Wang ◽  
Katarzyna Urbanska ◽  
Prannda Sharma ◽  
Mathilde Poussin ◽  
Reza Nejati ◽  
...  

Background: Peripheral T-cell lymphomas (PTCL) encompass a highly heterogeneous group of T-cell malignancies and are generally associated with a poor prognosis. Combination chemotherapy results in consistently poorer outcomes for T-cell lymphomas compared with B-cell lymphomas.1 There is an urgent clinical need to develop novel approaches to treatment of PTCL. While CD19- and CD20-directed immunotherapies have been successful in the treatment of B-cell malignancies, T-cell malignancies lack suitable immunotherapeutic targets. Brentuximab Vedotin, a CD30 antibody-drug conjugate, is not applicable to PTCL subtypes which do not express CD30.2 Broadly targeting pan-T cell markers is predicted to result in extensive T-cell depletion and clinically significant immune deficiency; therefore, a more tumor-specific antigen that primarily targets the malignant T-cell clone is needed. We reasoned that since malignant T cells are clonal and express the same T-cell receptor (TCR) in a given patient, and since the TCR β chain in human α/β TCRs can be grouped into 24 functional Vβ families targetable by monoclonal antibodies, immunotherapeutic targeting of TCR Vβ families would be an attractive strategy for the treatment of T-cell malignancies. Methods: We developed a flexible approach for targeting TCR Vβ families by engineering T cells to express a CD64 chimeric immune receptor (CD64-CIR), comprising a CD3ζ T cell signaling endodomain, CD28 costimulatory domain, and the high-affinity Fc gamma receptor I, CD64. T cells expressing CD64-CIR are predicted to be directed to tumor cells by Vβ-specific monoclonal antibodies that target tumor cell TCR, leading to T cell activation and induction of tumor cell death by T cell-mediated cytotoxicity. Results: This concept was first evaluated in vitro using cell lines. SupT1 T-cell lymphoblasts, which do not express a native functioning TCR, were stably transduced to express a Vβ12+ MART-1 specific TCR, resulting in a Vβ12 TCR expressing target T cell line.3 Vβ family specific cytolysis was confirmed by chromium release assays using co-culture of CD64 CIR transduced T cells with the engineered SupT1-Vβ12 cell line in the presence of Vβ12 monoclonal antibody. Percent specific lysis was calculated as (experimental - spontaneous lysis / maximal - spontaneous lysis) x 100. Controls using no antibody, Vβ8 antibody, and untransduced T cells did not show significant cytolysis (figure A). Next, the Jurkat T cell leukemic cell line, which expresses a native Vβ8 TCR, was used as targets in co-culture. Again, Vβ family target specific cytolysis was achieved in the presence of CD64 CIR T cells and Vβ8, but not Vβ12 control antibody. Having demonstrated Vβ family specific cytolysis in vitro using target T cell lines, we next evaluated TCR Vβ family targeting in vivo. Immunodeficient mice were injected with SupT1-Vβ12 or Jurkat T cells with the appropriate targeting Vβ antibody, and either CD64 CIR T cells or control untransduced T cells. The cell lines were transfected with firefly luciferase and tumor growth was measured by bioluminescence. The CD64 CIR T cells, but not untransduced T cells, in conjunction with the appropriate Vβ antibody, successfully controlled tumor growth (figure B). Our results provide proof-of-concept that TCR Vβ family specific T cell-mediated cytolysis is feasible, and informs the development of novel immunotherapies that target TCR Vβ families in T-cell malignancies. Unlike approaches that target pan-T cell antigens, this approach is not expected to cause substantial immune deficiency and could lead to a significant advance in the treatment of T-cell malignancies including PTCL. References 1. Coiffier B, Brousse N, Peuchmaur M, et al. Peripheral T-cell lymphomas have a worse prognosis than B-cell lymphomas: a prospective study of 361 immunophenotyped patients treated with the LNH-84 regimen. The GELA (Groupe d'Etude des Lymphomes Agressives). Ann Oncol Off J Eur Soc Med Oncol. 1990;1(1):45-50. 2. Horwitz SM, Advani RH, Bartlett NL, et al. Objective responses in relapsed T-cell lymphomas with single agent brentuximab vedotin. Blood. 2014;123(20):3095-3100. 3. Hughes MS, Yu YYL, Dudley ME, et al. Transfer of a TCR Gene Derived from a Patient with a Marked Antitumor Response Conveys Highly Active T-Cell Effector Functions. Hum Gene Ther. 2005;16(4):457-472. Figure Disclosures Schuster: Novartis, Genentech, Inc./ F. Hoffmann-La Roche: Research Funding; AlloGene, AstraZeneca, BeiGene, Genentech, Inc./ F. Hoffmann-La Roche, Juno/Celgene, Loxo Oncology, Nordic Nanovector, Novartis, Tessa Therapeutics: Consultancy, Honoraria.


1983 ◽  
Vol 158 (6) ◽  
pp. 2024-2039 ◽  
Author(s):  
M Howard ◽  
L Matis ◽  
T R Malek ◽  
E Shevach ◽  
W Kell ◽  
...  

Antigen-activated T lymphocytes produce within 24 h of stimulation a factor that is indistinguishable biochemically and functionally from the B cell co-stimulating growth factor, BCGF-I, originally identified in induced EL4 supernatants: Supernatants from antigen-stimulated T cell lines are not directly mitogenic for resting B cells, but synergize in an H-2-unrestricted manner with anti-Ig activated B cells to produce polyclonal proliferation but not antibody-forming-cell development; biochemical studies reveal the B cell co-stimulating factor present in antigen-stimulated T cell line supernatants is identical by phenyl Sepharose chromatography and isoelectric focusing (IEF) to EL4 supernatant BCGF-I. We thus conclude that normal T cells produce BCGF-I in response to antigenic stimulation. Analysis of the mechanism of BCGF-I production by antigen-stimulated T cells showed that optimum amounts of BCGF-I were obtained as quickly as 24 h post-stimulation, and that the factor producing cells in the T cell line investigated bore the Lyt-1+2- phenotype. As few as 10(4) T cells produced sufficient BCGF-I to support the proliferation of 5 X 10(4) purified anti-Ig activated B cells. Finally, the activation of normal T cell lines to produce BCGF-I required either antigen presented in the context of syngeneic antigen-presenting cells (APC) or interleukin 2 (IL-2).


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1345-1345
Author(s):  
Xingmin Feng ◽  
Tatsuya Chuhjo ◽  
Xuzhang Lu ◽  
Hiroyuki Takamatsu ◽  
Chiharu Sugimori ◽  
...  

Abstract A large body of evidence has suggested that acquired aplastic anemia (AA) of patients carrying HLA-DR15 is a kind of organ-specific autoimmune disease where hematopoietic progenitor cells in bone marrow are attacked by CD4+ T cells recognizing endogenous antigens. We recently identified diazepam-binding inhibitor-related protein 1 (DRS-1) as a candidate autoantigen capable of provoking immune system attack against hematopoietic progenitor cells in AA (Blood, 2004). Although in other organ-specific autoimmune diseases such as insulin-dependent diabetes mellitus and primary biliary cirrhosis, cytoplasmic proteins including glutamic acid decarboxylase 65 and pyruvate dehydrogenase complex have been shown to serve as autoantigens and mediate organ damages by CD4+ T cells, it remains unclear whether a peroxisomal protein like DRS-1 can be processed in hematopoietic progenitor cells, presented by HLA-DR15, and eventually serve as a target antigen of specific CD4+ T cells, leading to killing of hematopoietic progenitor cells themselves. To clarify these issues, we established a CD4+ T-cell line specific to a DRS-1 peptide (amino acid residues 191–204) from an AA patient carrying HLA-DR15 who had exhibited a high titer of anti-DRS-1 antibody as well as a high frequency of T-cell precursors specific to DRS-1, and then examined the cytotoxicity of the DRS-1-specific T-cell line against (1) autologous lymphoblastoid cell line (LCL) cells transfected with full length DRS-1 cDNA using a lentiviral vector, (2) myeloid leukemia cell lines carrying HLA-DR15 (KH88 and SAS413) and a leukemia cell line not carrying HLA-DR15 (K562), and (3) CD34+ progenitor cells from normal individuals. When all leukemia cell lines and LCL cells were examined for DRS-1 expression using Western blotting with specific monoclonal antibodies, DRS-1 protein was detected in DRS-1-transfected LCL cells, KH88 and K562, but not in nontransfected LCL cells and SAS413. Overexpression of DRS-1 gene by the CD34+ cells from normal individuals was ascertained by real-time PCR. In the 51Cr release assay, DRS-1-specific T cells showed cytotoxicity against only DRS-1-transfected LCL cells and KH88 in a dose-dependent manner (Figure), indicating that the T cell line requires presence of both DRS-1 and HLA-DR15 on target cells to exert cytotoxicity. When the DRS-1-specific T cells were incubated with CD34+ cells isolated from normal individuals with or without HLA-DR15 at an 10:1 ratio for 4 hours and cultured in a methylcellulose medium supplemented with colony-stimulating factors, the numbers of CFU-GM and BFU-E colonies derived from an HLA-DR15+ individual were 60.0% and 52.9% of a control whereas those derived from an HLA-DR15− individual were 90.1% and 88.2%. These findings indicate that hematopoietic progenitor cells in individuals with HLA-DR15 can present DRS-1 through the DR molecule and a breakdown of immune tolerance to DRS-1 may lead to development of AA.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3885-3885 ◽  
Author(s):  
Samantha Miner ◽  
Sawa Ito ◽  
Kazushi Tanimoto ◽  
Nancy F. Hensel ◽  
Fariba Chinian ◽  
...  

Abstract The immune-editing effect of myeloid leukemia has recently been reported in several studies. We previously demonstrated that the K562 leukemia-derived cell line suppresses T cell proliferation, which suggests that myeloid leukemia may function in a similar way to myeloid derived suppressor cells (MDSC). While the mechanism of suppression in leukemia is not fully understood, recent murine and human studies suggest that the STAT3 and arginase pathways play a key role in the immunosuppressive function of MDSC. We hypothesized that myeloid leukemia utilizes the MDSC STAT3 and arginase pathway to evade immune control, and block anti-leukemic immune responses. To evaluate the suppressive capacity of myeloid leukemia on T cell proliferation, we isolated CD34+ blasts and myeloid derived suppressor cells (MDSC: CD11b+CD14+) from blood of primary leukemia samples by FACS sorting (n=5). These cells were co-cultured with CFSE-labeled CD4+ T cells (n=9), previously isolated from healthy donor PBMCs using an automated cell separator (RoboSep). After stimulating with CD3/CD28 Dynabeads (Invitrogen, New York, USA) for 72 hours, proliferation was measured by CFSE dilution of the viable cell population. In three myeloid leukemias studied, CD4+ T cell proliferation was significantly suppressed in the presence of primary CD34 blasts and MDSC cells (p<0.001). Interestingly, CD34 blasts demonstrated a greater suppressive effect on T cells compared to MDSC cells for these samples (not statistically significant p=0.61). Next we repeated the proliferation assay using five leukemia cell lines: THP-1 and AML1 (derived from AML), K562 and CML1 (derived from CML), and the Daudi lymphoid-derived leukemia cell line. After staining with cell tracer dye and irradiating 100Gy, the cells were co-incubated with CFSE-labeled CD4+ T cells from healthy volunteers (n=6). We found that CD4+ T cell proliferation in the presence of the myeloid leukemia cell lines was significantly suppressed (mean proliferation 5.7±0.9% to 26.1±10.7%: p<0.0001 to 0.05) compared to lymphoid cell lines (mean proliferation 76.3±8.2%: p>0.05), consistent with the results obtained with the primary leukemia samples. To evaluate the impact of STAT3 and arginase on the immunosuppressive function of myeloid leukemia, the five cell lines were primed overnight with either arginase inhibitor (N(ω)-Hydroxy-nor-L-arginine; EMD Biosciences, Inc., California, USA) or two STAT3 inhibitors (STAT3 Inhibitor VI or Cucurbitacin I; EMD Millipore, Massachusetts, USA). Then, CD4+ T cells from healthy donors (n=3) were cultured with either (1) leukemia without any inhibitor (2) leukemia in the presence of inhibitor (3) leukemia primed with inhibitor. Priming leukemia with arginase inhibitor and STAT3 inhibitors almost completely abrogated their suppressive effect of T cell proliferation (p<0.001). We conclude that myeloid leukemia, like MDSC, directly immunosuppresses T cells, through STAT-3 and arginase. This finding may underlie the immune-editing of T cells by myeloid leukemia. Our results suggest that STAT3 inhibitors could be used to augment leukemia-targeted immunotherapy. Further investigation of T cell biology within the leukemia microenvironment is needed to further define immune editing mechanisms in myeloid leukemia. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures: No relevant conflicts of interest to declare.


1980 ◽  
Vol 152 (6) ◽  
pp. 1709-1719 ◽  
Author(s):  
S Gillis ◽  
J Watson

To isolate a stable tumor cell line capable of producing human interleukin 2 (IL-2; formerly referred to as T cell growth factor), 16 human T and B leukemia cell lines were screened for constitutive and mitogen-stimulated IL-2 production. We found that the T cell leukemia line designated Jurkat-FHCRC produced &gt; 200 U/ml of IL-2 activity after a 24-h stimulation with T cell mitogens. Peak mitogen-induced IL-2 activity was found in supernates harvested from 24-h Jurkat-FHCRC cell cultures stimulated with either 1% phytohemagglutinin or 20 microgram/ml concanavalin A. Addition of the fatty acid derivative phorbol myristate acetate to mitogen-stimulated cultures increased Jurkat-FHCRC IL-2 production to concentrations &gt; 400 U/ml. IL-2 activity observed in such cases represented between 100--300 times that produced in conventional cultures of mitogen- or alloantigen-stimulated normal human peripheral blood or splenic lymphocytes. Jurkat-FHCRC-derived conditioned medium demonstrated equal capacity to promote the sustained in vitro proliferation of either murine or human activated T cell lines confirming the ability of Jurkat-FHCRC cells to produce human IL-2. These studies identify a new source of human IL-2 and establish a valuable reagent for the isolation and further molecular characterization of this immunoregulatory molecule.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2537-2537
Author(s):  
Ashwini M Patil ◽  
Stefanie Kesper ◽  
Vishal Khairnar ◽  
Marco Luciani ◽  
Michael Möllmann ◽  
...  

Introduction: The thymus is a specialized hematopoietic organ, which is responsible for the generation of T cells. The central thymic cell type controlling T cell development are thymic epithelial cells (TECs). Based on their specific function and anatomic location TECs are separated into cortical and medullary subsets (cTECs and mTECs). cTECs express pivotal NOTCH-ligands such as DLL4 controlling T cell lineage commitment while mTECs play a central role in negative selection of developing T cells. Acquisition of NOTCH1 gain-of-function mutations play a central role in acute T cell lymphoblastic leukemia (T-ALL) development. During T-ALL leukemogenesis aberrant expression of transcription factors such as SCL/TAL1 and LMO1 block T cell differentiation and increase self-renewal while NOTCH1 mutations promote survival and proliferation. Since most acquired NOTCH1 mutations still require ligand binding to exert augmented signaling we propose DLL4-expressing TECs playing a critical role during T-ALL leukemogenesis. Methods: In the present study, we used a Scl/Lmo1 T-ALL transgenic mouse model, murine ANV and TE71 TEC cell lines and human T-ALL cell lines (Jurkat, ALL-SIL, DND-41, and HPB-ALL) to investigate TEC dynamics and function in the T-ALL context. Results: First, we demonstrated T-ALL supporting potential of TEC cell lines in vitro, which was comparable to the mesenchymal cell line OP9. Next, we showed in the Scl/Lmo1 T-ALL mouse model which had a mean survival rate of 90 days that preleukemic thymocytes displayed a striking upregulation of Notch1 target genes. Interestingly, fluorescence microscopy revealed a relative expansion of cortical and a relative reduction of the medullary thymic areas in Scl/Lmo1 thymi (Fig. 1A). Correspondingly, absolute numbers of cTECs expanded while mTEC numbers declined (Fig. 1B). Gene expression profiling of sorted preleukemic Scl/Lmo1 cTECs revealed upregulation of the chemokine CXCL10 (Fig. 1C). Moreover, increased CXCL10 chemokine concentrations were detected in Scl/Lmo1 thymic interstitial fluid (Fig.1D). Strikingly, we demonstrated T-ALL dependence of TEC Cxcl10 upregulation. We showed that Cxcl10 upregulation in TEC cell lines was only induced by direct cellular contact with Scl/Lmo1 thymocytes while wild-type control thymocytes did not alter TEC cell line Cxcl10 expression (Fig. 1E). Next, a high proportion of the CXCL10 receptor CXCR3 expressing cells was revealed in Scl/Lmo1 thymi (Fig. 1F) and by human T-ALL cell lines. Finally, we demonstrated a CXCL10 dependent pro-survival effect within cultured SCL/LMO1 thymocytes (Fig. 1G), which was associated with the activation of NOTCH1 signaling (Fig. 1H). Conclusions: In summary, the data support a novel T-ALL-promoting regulatory circuit in which emerging T-ALL lymphoblasts induce CXCL10 in expanding TECs which positively feeds back to T-ALL cells via the CXCL10 receptor CXCR3. Disclosures Dührsen: Celgene: Research Funding; Takeda: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Research Funding; Teva: Honoraria; Novartis: Consultancy, Honoraria; Alexion: Honoraria; Roche: Honoraria, Research Funding; CPT: Consultancy, Honoraria; Janssen: Honoraria. Göthert:Proteros Biostructures: Consultancy; Novartis: Consultancy, Honoraria, Other: Travel support; Pfizer: Consultancy, Honoraria; Incyte: Consultancy, Honoraria, Other: Travel support; Bristol-Myers Squibb: Consultancy, Honoraria, Other: Travel support; AOP Orphan Pharmaceuticals: Honoraria, Other: Travel support.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1849-1849
Author(s):  
M. Kozik ◽  
M.L. Lesniewski ◽  
Y. Hegerfeldt ◽  
R.R. Brewka ◽  
L.R. Fanning ◽  
...  

Abstract Introduction: NFAT1 is a transcription factor integral for the regulation of T-cell proliferation, differentiation, and apoptosis. NFAT1 is present in the cytoplasm of resting T-cells and upon stimulation (ionomycin, antigen or anti-CD3) is dephosphorylated via calcineurin and translocates to the nucleus to associate with other transcription factors (AP1). We sought to determine whether NFAT1 regulation in T-cells contributes to leukemogenesis. Initial in vitro analyses incorporated several T-ALL cell lines all derived from the peripheral blood of patients including 3 mature T-cell lines: CCRF-CEM (no clear chromosomal abnormalities), Jurkat (karyotype 46, XY, −2, −18, del(2) (p21p23), del(18) (p11.2)) Loucy (translocation t(16;20)(p12;q13), and p53 overexpression), and one immature T-cell line Molt-4 (hypertetraploid chromosomes and 6q-, t(7;7)) (ATCC Manassas, VA). Methods: T-ALL cell lines were cultured in accordance with ATCC guidelines. Surface marker analysis for CD34, CD38, HLA-DR, CD3, CD4, CD8, CD2, and CD7 was performed (BD Biosciences). Cytoplasmic and nuclear extracts were prepared from cell lines and control adult blood (AB) CD3+ cells. Cell lysates (20μg) were examined by Western blot. Blots were probed with anti-NFAT1 antibody (BD Biosciences) and protein loading controls β2-microglobulin (Abcam) and lamin A/C (Santa Cruz Biotechnology). Bands were visualized using Supersignal West Pico chemiluminescent substrate (Pierce) and quantitated using NIH imageJ software. Apoptotic assays were performed by treating samples with 0.832 μM cyclosporin A (CsA) for 48 hrs, then determining the percentage of early apoptotic cells using an Annexin V/Propidium iodide flow cytometric kit (Calbiochem). Results: Surface marker analysis confirmed that Loucy, Jurkat, and CCRF-CEM cells possess a mature phenotype, whereas Molt-4 cells possess a more immature phenotype (Table 1). Western blot analysis showed Loucy cells were unique in that NFAT1 was absent in the nuclear fraction, indicating NFAT1 is only present in it’s inactive form in this cell line. Among the T-ALL cell lines, only Loucy showed no difference in the proportion of apoptotic cells following CsA treatment. Table 1. Phenotype Analysis of T-ALL Cell Lines Surface Antigens CCRF-CEM JURKAT LOUCY MOLT-4 Percentage of Total Cells CD2 83.97 96.16 4.96 91.53 CD7 99.74 87.84 99.96 57.88 CD3 18.22 52.15 96.48 8.33 CD4 62.42 54.56 12.62 42.64 CD8 97.42 5.32 0.81 83.19 CD34 14.80 2.62 0.65 55.78 CD38 93.59 89.40 92.92 99.66 HLA-DR 2.31 2.62 0.56 0.33 Conclusions: Despite Loucy cells’ mature phenotype, they exhibited distinct NFAT1 dysregulation compared to other mature cell lines. CsA treatment of Loucy cells failed to effect NFAT1 translocation to the nucleus and induction of apoptosis, as NFAT1 remained in the cytoplasm. Loucy’s insensitivity to CsA with regards to apoptosis signaling supports the idea for a role for NFAT1 in Ca(2+) signaling cascade for apoptosis in T-cells. These results also suggest a possible role of cytosolic NFAT1 in leukemiogenesis, since the amount of protein seen in whole cell extracts between the various cell lines was nearly the same. The translocation in Loucy cells is in the same chromosomal location as the NFAT1 gene locus. Studies are ongoing to understand the role of NFAT1 in T-ALL initiation and progression.


Sign in / Sign up

Export Citation Format

Share Document