KLF4 induces apoptosis in T-ALL through the BCL2/BCLXL pathway

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4902-4902
Author(s):  
Bing Xu ◽  
Xiangmeng Wang ◽  
Peng Li ◽  
Wei Li ◽  
Huijuan Dong ◽  
...  

Abstract KLF4, also known as GKLF (gut KLF), is a member of the KLF zinc finger-containing transcription factor family. Klf4 together with Oct4, Sox2, and c-Mycare widely referred to as ‘Yamanaka factors’ because mouse somatic cells can be reprogrammed into pluripotent stem cells following their ectopic expression. The transcription factor Kruppel-like factor 4 (KLF4) may induce tumorigenesis or suppress tumor growth in a tissue-dependent manner. In T cell leukemia and pre-B cell lymphoma cells, KLF4 acts as a tumor suppressor. We found that over expression of KLF4 induced human acute T cell lymphoblastic leukemia (T-ALL) cell lines to undergo apoptosis through the BCL2/BCLXL pathway, and we confirmed KLF4-induced apoptosis in primary samples from T-ALL patients. We further characterized KLF4 function in human early and mature T cells. Our analysis uncovered that KLF4 suppressed the transcription of other T cell-associated genes in T-ALL. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3325-3325
Author(s):  
Mariska T. Meijer ◽  
Valentina Cordo' ◽  
Rico Hagelaar ◽  
Willem K. Smits ◽  
Jules PP Meijerink

Abstract Ectopic MEF2C expression due to rearrangements have been recurrently found in patients with human early thymocyte progenitor acute lymphoblastic leukemia (ETP-ALL). ETP-ALL is the most immature subtype of T-ALL and is characterized by the expression of myeloid lineage markers and an ETP-like gene expression profile. In normal haematopoiesis, MEF2C expression occurs at immature stages and in myeloid- and B cell lineages but is absent in early T cell progenitors. Here, we investigate MEF2C as an oncogene for ETP-ALL. First, we validated our initial observations in an independent T-ALL cohort that ETP-ALL patients highly express MEF2C along with BCL2, HHEX, LMO2 and LYL1 in contrast to other T-ALL subtypes. In addition, we have demonstrated that enforced MEF2C expression in the T-lineage drives a bi-phenotypic CD3 +CD19 + leukemia in mice by inhibiting a T-cell developmental program in favor a B-cell transcriptional program and that resembles human ETP-ALL. This confirms MEF2C as an oncogenic driver of ETP-ALL. We developed a model system that enabled screening of various drug libraries for compounds that overcome developmental arrest induced by MEF2C-expression, as evidenced by the upregulation of CD3 and TCRγδ. The ETP-ALL-like LOUCY cells express MEF2C as consequence of a del5(q14-qter). Further induction of MEF2C levels (i.e. LOUCY-iMEF2C cells) blocked differentiation of LOUCY cells towards a CD3 +/TCRγδ + phenotype on OP9-DL1 or DLL4-coated plates in contrast to parental LOUCY cells. This model enabled high-throughput screening of drug libraries using a flow cytometry-based assay for drugs that further promote differentiation of LOUCY-MEF2C cells. We identified various epigenetic inhibitors that promoted differentiation of LOUCY-iMEF2C cells. Unlike AML patients, where HDAC4 was shown to play an important role in the activation of MEF2C and the propagation of the leukemia, we found that different HDACs are involved that can block or promote MEF2C function. This suggests that the role of different HDACs in MEF2C regulation is cell-type or disease dependent. In addition, over-expression of MEF2C raised cellular resistance towards prednisolone treatment. Conversely, inhibition of MEF2C sensitized cells to prednisolone cytotoxicity, even in the presence of MEF2C-overexpression. Together, these data show that MEF2C is an oncogenic driver of ETP-ALL, and ectopic expression of MEF2C negatively affects steroid sensitivity. Specific targeting of only those HDACs that promote MEF2C activity may provide new therapeutic options in ETP-ALL by inhibiting MEF2C function and enhancing steroid sensitivity. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1026-1026
Author(s):  
Erica A. Lehotzky ◽  
Mark Y. Chiang

Abstract Abstract 1026 Despite numerous advances in the past few decades, treatment of acute lymphoblastic leukemia/lymphoma (ALL) remains a common and considerable challenge. Further efforts to define the molecular lesions that drive ALL are needed to improve clinical management. The Hox subfamily of T-cell ALL (T-ALL) represents 30–40% of pediatric and adult cases. TLX1/HOX11 is the prototypical member of the Hox group. To generate a resource for developing targeted therapies for Hox T-ALLs, we developed a doxycycline-regulated mouse model of Tlx1-initiated T-ALL. Dysregulated thymic expression of Tlx1 induces T-ALL after ∼5-7 months with penetrance of 15–60%. The lymphoblasts are arrested at the early CD4+/CD8+/CD24hi stage of T-cell development, similar to human T-ALLs of the TLX1 subtype. Spontaneous activation of the Notch1 oncogene occurred in the tumors. In about two-thirds of samples, Notch was activated through acquired mutations in the heterodimerization and PEST domains that resemble the Notch1 mutations found in human patients. Inhibition of Notch signaling with g-secretase inhibitors completely abrogated cell line growth and induced apoptosis. Notch inhibition also transiently delayed leukemia progression by ∼17 days in vivo. In contrast, suppression of Tlx1 expression had more moderate inhibitory effects on cell line growth in vitro. However, suppression of Tlx1 expression in transgenic mice transiently delayed leukemia progression by ∼11 days. Tlx1 suppression had the strongest inhibitory effects on expression of CCR7 and lymph node size. These effects were fully reversed with ectopic expression of Tlx1. These data show that Tlx1 can convert normal thymocytes into leukemia cells, but the leukemia cells are not fully dependent on continued Tlx1 expression. The leukemia cells recruit secondary factors and pathways such as Notch and c-Myc to sustain growth and survival. Our study highlights a strong resiliency of T-ALL cells to both Tlx1 and Notch inhibition. Our study has important implications for targeting these pathways for the treatment of T-ALL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2729-2729
Author(s):  
Yuan-I Chang ◽  
Guangyao Kong ◽  
Jing Zhang ◽  
Erik A. Ranheim

Abstract Recent whole genome/exome sequencing efforts in myeloid malignancies identified that mutations in DNA methyltransferase 3A (DNMT3A) are prevalent in acute myeloid leukemia (AML). In addition, DNMT3A mutations are also identified in various T cell malignancies. Of note, DNMT3A mutations are typically heterozygous and some WT DNMT3A functions thus remain in this state. However, the predominant DNMT3A R882 mutations, which locate in the catalytic domain, seem to inhibit the methyltransferase activity of the remaining WT DNMT3A due to its dominant-negative function (Yang L, Rau R, Goodell MA, Nat. Rev. Cancer 15: 152-165, 2015). COSMIC database analysis reveals different prevalence of DNMT3A R882 mutations in various hematopoietic malignancies. Approximately 60% of DNMT3A mutations in AML are R882 mutations, while the frequency of R882 mutations drops to ~40% in myelodysplastic syndrome (MDS) and myeloproliferative neoplasm (MPN). In contrast, the frequency of R882mutations is less than 25% in T-cell acute lymphoblastic leukemia (T-ALL). The significantly different frequencies of DNMT3A R882 mutations in AML versus T-ALL inspired us to investigate whether downregulation of DNMT3A regulates malignancies of different lineages in a dose-dependent manner. We previously showed that Dnmt3a-/- promotes MPN progression in KrasG12D/+ mice and ~1/3 compound mice develop AML-like disease (Chang et al. Leukemia 29: 1847-1856, 2015). Here, we generated KrasG12D/+; Dnmt3afl/+; Mx1-Cre mice to determine how Dnmt3a haploisufficiency affects KrasG12D/+-induced leukemogenesis. After pI-pC injections to induce Mx1-Cre expression, primary KrasG12D/+; Dnmt3a+/- mice died quickly as primary KrasG12D/+ mice; the survival rates of these two groups of animals were not significantly different. However, in a competitive transplant setting, recipients transplanted with KrasG12D/+; Dnmt3a+/- bone marrow cells displayed a significantly shortened survival than recipients with KrasG12D/+ cells. Moreover, all of the recipients with KrasG12D/+; Dnmt3a+/- cells developed a lethal T-ALL without significant MPN phenotypes, while ~20% of recipients with KrasG12D/+ cells developed MPN with or without T-ALL. This is in sharp contrast to the recipients with KrasG12D/+; Dnmt3a-/- cells, in which ~60% developed a lethal myeloid malignancy (MPN or AML). Our data suggest that in the context of oncogenic Kras, loss of Dnmt3a promotes myeloid malignancies, while Dnmt3a haploisufficiency induces T-ALL. This dose-dependent phenotype is highly consistent with the prevalence of DNMT3A R882 mutations in AML versus T-ALL in human. We are currently investigating the underlying mechanisms. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2641-2641
Author(s):  
Suning Chen ◽  
Stefan Nagel ◽  
Bjoern Schneider ◽  
Maren Kaufmann ◽  
Ursula R. Kees ◽  
...  

Abstract Abstract 2641 Poster Board II-617 Background: In T-cell acute lymphoblastic leukemia (T-ALL) the LMO2 transcription factor locus is juxtaposed with T-cell receptor (TCR) genes by a recurrent chromosome translocation, t(11;14)(p13;q11). Recent molecular cytogenetic data indicate that unlike classical TCR rearrangements, t(11;14) operates synonymously with submicroscopic del(11)(p13p13) by removing a negative upstream LMO2 regulator (Dik et al., Blood 2007;110:388). The combined incidence of both LMO2 rearrangements is ∼10-15% (Van Vlierberghe and Huret, Atlas Genet Cytogenet Oncol Haematol, November 2007). However, aberrant LMO2 expression occurs in nearly half of all T-ALL cases, a discrepancy which may indicate a significant contribution by cryptic chromosome alterations. We attempted the extended characterization of the LMO2 genomic region in T-ALL cell lines to look for such rearrangements. Cells and Methods: We investigated a panel of 26 well characterized and authenticated T-ALL cell lines using parallel fluorescence in situ hybridization (FISH) with a tilepath BAC/fosmid contig and both conventional and quantitative reverse transcriptase (Rq)-PCR. Global gene expression was additionally measured in some cell lines by Affymetrix array profiling. Results: LMO2 rearrangements were detected in 5/26 (19.2%) cell lines including both established rearrangements, t(11;14) and del(11)(p13p13) in one cell line apiece (3.8%). Interestingly, we found two novel LMO2 translocations: t(X;11)(q25;p13) in 2/26 (7.7%), and t(3;11)(q25;p13) in 1/26 (3.8%) cell lines, respectively. Comparing transcription levels in cell lines with and without genomic rearrangements showed that LMO2 expression was significantly higher in T-ALL cell lines carrying LMO2 rearrangements (P<0.001). Rq-PCR revealed that 5 of the top 10 (50%) LMO2 expressing cell lines carry cytogenetic rearrangements at this locus, compared to 0/16 remaining examples. Loss of a recently defined LMO2 negative regulatory element was identified in the del(11)(p13p13) cell line but no other deletions were detected. Two genes STAG2 at Xq25 and MBNL1 at 3q25 were identified as novice LMO2 partners in t(X;11) and t(3;11), respectively. In both genes breakpoints lay at intron 1 close to deeply conserved noncoding regulatory regions. Both t(X;11) cell lines displayed conspicuous silencing of the ubiquitously expressed STAG2 gene highlighting the transcriptional significance of the region displaced. Unlike t(11;14)/del(11)(p13p13) both new rearrangements carry LMO2 breakpoints in the far upstream region (at minus 80–150 Kbp), and appear to result in upregulation of LMO2 by juxtaposition rather than via covert deletion. STAG2 is a component of the chromosomal cohesin complex which acts as a transcriptional coactivator, and which has been recently identified as a potential driver of oncogene transcription in acute myeloid leukemia (Walter et al., Proc Natl Acad Sci U S A. 2009;106:1295). MBNL1 controls RNA splicing and is a rare BCL6 partner gene in B-cell lymphoma, but this is the first report of its involvement in T-ALL. Conclusion: Given their frequency and variety in a small sample, we propose that cryptic chromosome rearrangements targeting LMO2 upregulation may be significantly more frequent than hitherto appreciated in T-ALL. Unlike canonical LMO2 rearrangements, both t(X;11) and t(3;11) would appear to function positively by upregulation of LMO2 via juxtaposition with noncoding driver elements within these novel partner genes. Perspectives: Future work will address the regulatory potential of candidate enhancer sequences embedded within conserved noncoding intronic sequences of MBNL1 and STAG2. Cytogenetically inconspicuous cell lines displaying LMO2 upregulation will be subjected to more detailed scrutiny using high density genomic SNP arrays. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 9-9
Author(s):  
Irene Homminga ◽  
Rob Pieters ◽  
Anton Langerak ◽  
Johan de Rooi ◽  
Andrew Stubbs ◽  
...  

Abstract Abstract 9 To identify novel oncogenic pathways in T-cell acute lymphoblastic leukemia (T-ALL), we combined expression profiling of 117 pediatric patient samples and detailed molecular cytogenetic analyses. Using unsupervised and supervised analyses, we identified a T-ALL cluster that was associated with an immature immunophenotype (CD1−, CD4−, CD8−), frequent expression of CD34 and co-expression of the myeloid markers CD13/CD33. Patients in this cluster lacked any of the known oncogenic rearrangements, but ectopically expressed MEF2C, which was recently demonstrated as an important transcription factor for T-cell development1. Molecular-cytogenetic analyses including the Chromatine Conformation Capture on Chip (4C) method revealed novel rearrangements of the MEF2C locus at 5q14, rearrangement of transcription factors that target MEF2C (PU.1, NKX2-5, RUNX1) or MEF2C-associated cofactors (NCOA2/GRIP1) in about half of the patients in this cluster. Four out of the 6 rearrangements identified have never been observed before in human cancer. Nearly all of these patients in this cluster could be predicted by the early T-cell precursor (ETP) signature2 using PAM statistics. This indicates that MEF2C may represent the oncogene for ETP T-ALL, an entity that has been associated with poor outcome2. Inhibition of MEF2C in a cell line model system provoked relieve of developmental arrest, indicating that ectopic MEF2C expression blocks T-cell development at an early stage. We demonstrated that MEF2C is a transcriptional regulator for many differentially expressed genes that were associated with the immature cluster including LYL1 and LMO2. Although LYL1 has been suggested as potential oncogene for immature T-ALL cases3, oncogenic rearrangements were never identified in T-ALL cases with immature immunophenotype. Our data therefore imply that high expression of LYL1 (and LMO2) is part of a pathogenic pathway for immature T-ALL that is regulated by the MEF2C oncogene. 1 Stehling-Sun, S., Dade, J., Nutt, S. L., DeKoter, R. P. & Camargo, F. D. Regulation of lymphoid versus myeloid fate ’choice’ by the transcription factor Mef2c. Nat Immunol 10, 289–296, (2009). 2 Coustan-Smith, E. et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol 10, 147–156, (2009). 3 Ferrando, A. A. et al. Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer Cell 1, 75–87 (2002). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2405-2405
Author(s):  
Bing Xu ◽  
Peng Li

Abstract The transcription factor Kruppel-like factor 4 (KLF4) may induce tumorigenesis or suppress tumor growth in a tissue-dependent manner. We found that overexpression of KLF4 induced not only human acute T-acute lymphoblastic leukemia (T-ALL) cell lines but also primary samples from T-ALL patients to undergo apoptosis through the BCL2/BCLXL pathway in vitro. T cell-associated genes including BCL11B, GATA3, and TCF7 were negatively regulated by KLF4 overexpression. Especially, KLF4 induced SUMOylation and degradation of BCL11B. However, the KLF4-induced apoptosis in T-ALL was rescued by the in vivo microenvironment. Furthermore, the invasion capacity of T-ALL to hosts was compromised when KLF4 was overexpressed. In normal human T cells, the overexpression of KLF4 severely impaired T cell development at early stages, but the blockage of T cell development was resumed by restoration of GATA3 or ICN1. In summary, our data demonstrate that KLF4 acts as a tumor suppressor in malignant T cells and that downregulation of KLF4 may be a prerequisite for early human T cell development and homeostasis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1136-1136
Author(s):  
André De Almeida ◽  
Tim Pieters ◽  
Sara T'Sas ◽  
Steven Goossens ◽  
Pieter Van Vlierberghe

Abstract Background: T-lineage acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy that accounts for 10%-15% of pediatric and 25% of adult ALL cases. Although the prognosis of T-ALL has been improving over time, the outcome of T- ALL patients with primary resistant or relapsed leukemia remains poor. Thus, further advances in the treatment of T-ALL require the identification of new targets for the development of highly specific molecularly targeted drugs. SRY-related HMG-box 11 (SOX11) is a member of the SOXC family, which also includes SOX4 and SOX12. SOX11 is single-exon gene that encodes a transcription factor, but its molecular properties and functions remain incompletely understood. Expression of SOX11 is strongly associated with neurogenic activity, and it is confined to the central nervous system. Aberrant SOX11 upregulation has been observed in several cancer types, such as medulloblastoma, glioma, and B- and T-cell lymphomas. Under physiological conditions, SOX11 is absent in the T-cell lineage. However, SOX11 expression is consistently found in a subset of TAL/LMO T-ALL cases characterized by an abundancy of PTEN mutations and an underrepresentation of NOTCH mutations. Since the expression of SOX11 is restricted to a malignant context, we postulate that SOX11 might act as an oncogene in T-ALL. Aims: We want to assess SOX11's potential to promote a (pre)leukemic state, and to identify the role of SOX11 during oncogenic transformation in vivo. Methods: We generated a R26-SOX11 knock-in mouse model that allows conditional SOX11 expression in thymocytes, by using Lck-Cre (SL). Using this model, we analyzed the (immuno)phenotype and transcriptome of T-cell populations in 8- to 12-weeks old mice. In the future, we will also perform a syngeneic transplantation of SL thymocytes into sublethally irradiated mice to assess their colonizing potential. Additionally, we have set up an aging cohort of SL mice to investigate whether expression of SOX11 suffices to induce malignant transformation. Since the expression of SOX11 is associated with the TAL/LMO subgroup, we also crossed SL mice with a well-established murine T-ALL model, CD2-Lmo2, to give rise to mice that express both transgenes in their thymocytes (SCL). An aging cohort of this T-ALL model was followed, and we generated a survival curve and collected biological samples. These tumor samples are being analyzed, and their transcriptomic profile will be compared with that of mice overexpressing Lmo2 only. Lastly, we will lentivirally transduce human T-ALL cell lines from the TAL/LMO subgroup using a vector for SOX11 expression. These cell-lines will be used to study the consequences of expressing this transcription factor in a pre-established malignant setting. Results: In order to determine the effects of SOX11 expression in T-cells, we collected and analyzed blood, spleen, and thymus of 12-weeks old SL mice and their Cre-negative littermates (WT). To validate this model, we performed real-time PCR and immunoblotting to confirm the overexpression of SOX11 in T-cells. Interestingly, we observed a significant reduction in the number of lymphocytes in the peripheral blood and in the spleen of SL mice. Furthermore, significant changes were found in the proportions of all thymic populations, featuring an increase in double negative (DN) T-cells (Fig. 1A), particularly the DN3 subpopulation (Fig. 1B), and a decreased double positive CD4/CD8 fraction (DP). Based on that finding, we decided to sort out DN3 and DP cells from WT and SL thymi, and performed RNA-sequencing to further characterize the effect of SOX11 in these cells. In addition, we are following a cohort of SL mice, but so far no tumor formation was observed (average age of the cohort=300-days old). On the other hand, simultaneous overexpression of SOX11 and Lmo2 greatly accelerated the development of T-cell malignancies in SCL mice when compared with CD2-Lmo2 mice (Fig. 1C; median survival of 177 days versus 307; p&lt;0.0001), revealing a synergistic effect upon combination of both genetic events. Not surprisingly, most tumors were composed of immature T-cells, mainly DN cells, and no noteworthy differences were found between the composition and location of SCL and CD2-Lmo2 tumors. Conclusion: We showed that SOX11 skews the normal development of thymocytes in vivo. Additionally, we developed a murine SOX11 T-ALL model, thus confirming an oncogenic role for SOX11. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3636-3636
Author(s):  
Stefan Nagel ◽  
Letizia Venturini ◽  
Corinna Meyer ◽  
Maren Kaufmann ◽  
Michaela Scherr ◽  
...  

Abstract Abstract 3636 Myocyte enhancer factor 2C (MEF2C) is a transcription factor of the MADS-box family which is physiologically expressed in hematopoietic stem cells and during development of B-cells. Recently, we identified ectopic expression of MEF2C in T-cell acute lymphoblastic leukemia (T-ALL) cell lines activated either via chromosomally mediated ectopic expression of homeodomain protein NKX2-5 or via deletion of non-coding exon and promoter regions at 5q14, suggesting loss of negative regulatory elements. Our aim was to identify additional transcriptional regulators of MEF2C in T-ALL. Therefore, we analyzed the sequence of the MEF2C 5′-region, thus identifying potential regulatory binding sites for GFI1B, basic helix-loop-helix (bHLH) proteins, STAT5 and HOXA9/HOXA10. Overexpression studies demonstrated MEF2C activation by GFI1B (strong), LYL1 and TAL1 leukemic bHLH proteins (weak), and inhibition by STAT5 (strong) and HOXA9/HOXA10 (weak). Chromatin-Immuno-Precipitation analysis demonstrated direct binding of GFI1B, LYL1 and STAT5 but not of HOXA10 to the MEF2C 5′-region in T-ALL cells. However, HOXA9/HOXA10 activated expression of NMYC which in turn mediated MEF2C repression, indicating an indirect mode of MEF2C regulation. Chromosomal deletion of the 5′-MEF2C STAT5 binding site in LOUCY cells by del(5)(q14), reduced expression levels of STAT5 protein in some MEF2C-positve T-ALL cell lines, and the presence of inhibitory IL7-JAK-STAT5-signaling highlighted the repressive impact of this factor in MEF2C regulation. Taken together, our results indicate that ectopic expression of MEF2C in T-ALL cells is mainly regulated via activating leukemic transcription factors GFI1B or NKX2-5 and by escaping inhibitory STAT5-signaling. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1463-1463
Author(s):  
Kimberly G Laffey ◽  
Robert J Stiles ◽  
Melissa Ludescher ◽  
Tessa Davis ◽  
Shariq S Khwaja ◽  
...  

T cell lymphoblastic leukemia (T-ALL) is an aggressive cancer arising from transformed thymocytes. Most human T-ALL involves hyperactive NOTCH signaling that is often caused by activating NOTCH mutations. However, the identification of specific molecular signals that might induce or select for mutation and transformation are incompletely understood. We report that an understudied low-frequency, natural thymocyte subset expresses αβ T cell antigen receptor (TCR) earlier than most cells in mice and humans; engagement of the early αβTCR by major histocompatibility complexes (MHC) can cause outgrowth of NOTCH1 mutant clones and T-ALL leukemogenesis in a mouse model of T-ALL. Assessment of 5 recent human T-ALL cases found one to present this unique CD4-CD8- double- negative (DN) stage as the earliest identifiable developmental stage. These studies present a model of T-ALL leukemogenesis that identifies (i) a natural cell stage of origin susceptible to transformation, (ii) a matching mouse model showing that a signaling receptor (αβTCR) and its ligand (MHC) drive leukemogeneis and outgrowth of tumors bearing activating NOTCH1 mutations, and (iii) a human case that presents with a tumor consistent with this model and mechanism. In past work, the pre-TCR has been shown to impact T-ALL development in mice (Campese et al, Blood 2006), but an oncogenic role for the mature αβTCR is less well characterized and somewhat surprising. This is because, although T-ALL tumor cells may express variable levels of surface αβTCR/CD3, the earliest cell stages that are thought to transform are also thought to precede stages with αβTCR expression. Most conventional αβ thymocytes rearrange TCRβ and TCRα loci in separate, ordered developmental stages. However, some thymocytes in the conventional pathway rearrange both at DN stage thus exhibiting 'precocious' αβTCR (PAT) expression. Importantly, these PAT cells are indeed part of the conventional αβ lineage, being a 'subset' only due to early αβTCR expression but without known distinction in ultimate immune function (Aifantis et al, JEM 2006). We found that ~0.01% of mouse and human thymocytes are such PAT cells at steady state. To interrogate the PAT thymocyte surface phenotype, we performed multi-parametric flow cytometry with Spanning-tree Progression Analysis of Density-normalized Events (SPADE). This revealed that PAT thymocytes constitute a DN subset that is not associated with other well-described unconventional DN thymocytes known to express αβTCR, consistent with as the expectation that PATs are part of the conventional developmental pathway. We observed that the OT1 TCR transgene is expressed in mice with parallel timing and level to the natural PAT subset, allowing use of this model to study antigen-dependent signaling and oncogenesis. In a cohort study, no T-ALL was observed in wild-type C57BL/6 or OT-1.β2M-/- mice (deficient in endogenous antigen presentation), but MHC-sufficient OT-1 mice developed PAT-stage-specific T-ALL with activating NOTCH1 mutations. Transplant experiments corroborated a requirement for antigen presentation and TCR signaling for tumor maintenance as transplanted tumors grew in MHC+ but not MHC-deficient mice. This predicted that PAT thymocytes might have an unusual ability to signal through αβTCR even without coreceptor expression. When cultured in the presence of either exogenously added β2M or antigen presenting cells, both untransformed and neoplastic PAT cells upregulated CD69 in response to the OT-1 antigenic peptide, OVA. Furthermore, ex vivo analysis of PAT cells from polyclonal C57BL/6 versus MHC-deficient mice showed intrinsic upregulation of TCR-signaling-dependent Nur77 in an MHC-dependent manner. These data revealed a unique ability of PAT cells to engage in co-receptor independent but antigen-dependent signaling. Microarray analysis showed that the gene expression profile of neoplastic PAT cells from OT-1 T-ALL most closely resembled that of conventional post β-selection DN thymocytes, in agreement with the natural PAT stage during normal T cell development. These data support a model in which transformation occurred in the naturally occurring αβ PAT thymocyte subset as cell-of-origin. Collectively, our data suggest that precocious αβTCR expression and coreceptor-independent antigen engagement can cause activating NOTCH mutation and T-ALL development. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4811-4811 ◽  
Author(s):  
Michael W Harr ◽  
Paolo F. Caimi ◽  
Karen McColl ◽  
Fei Zhong ◽  
Shilen N Patel ◽  
...  

Abstract Abstract 4811 Glucocorticoid hormones are frequently used as part of front-line therapy to treat lymphoid neoplasms. However, relative to acute lymphoblastic leukemia and many types of B cell lymphoma, chronic lymphocytic leukemia (CLL) cells are intrinsically resistant to glucocorticoids and the mechanism governing this resistance has not been fully investigated. Here we report that that the Src inhibitor dasatinib enhances glucocorticoid-induced apoptosis in lymphoid cells and sensitizes primary CLL cells to dexamethasone. This finding evolved from the observation that the Src kinase Lck was downregulated by dexamethasone in glucocorticoid sensitive T cells in order to suppress immunoreceptor activation and signaling. In contrast, aberrantly expressed Lck was not downregulated by dexamethasone in CLL cells. However, inhibition of Lck phosphorylation by dasatinib significantly enhanced cell killing to nanomolar concentrations of dexamethasone. Collectively, these data indicate that dasatinib sensitizes primary CLL cells to glucocorticoids, and thus, the combination of these agents may have therapeutic efficacy in patients with aggressive or refractory CLL. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document