PAX5-Mediated Lineage Conversion and Expression of AID Accelerates Clonal Evolution and Initiates Darwinian Selection of BCR-ABL1-Mutants in Chronic Myeloid Leukemia.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1005-1005 ◽  
Author(s):  
Lars Klemm ◽  
Niklas Feldhahn ◽  
Thomas K. Hoffmann ◽  
Wolf-Karsten Hofmann ◽  
Hassan Jumaa ◽  
...  

Abstract Chronic Myeloid Leukemia (CML) is typically derived from the myeloid lineage. CML cells in terminal blast crisis, however, often exhibit a B lymphoid phenotype. The reasons for progression into blast crisis and myeloid/B lymphoid lineage switch are largely unknown. Studying expression of the B cell-specific mutator enzyme AID in BCR-ABL1-transformed leukemias, we found that AID is expressed in BCR-ABL1-positive B cell lineage ALL and B lymphoid blast crisis CML but not chronic phase or myeloid blast crisis CML. Studying five primary cases of mixed lineage bast crisis CML, AID expression was only found in the sorted B cell lineage but not myeloid lineage subclones. This pattern of AID expression correlated with a high frequency of DNA single-strand breaks within the tumor suppressor genes CDKN2A and CDKN2B, which were found in B cell lineage but not myeloid lineage subclones of blast crisis CML. Interestingly, the expression pattern or AID parallels the frequency of mutations within the BCR-ABL1 kinase domain that confer resistance to the BCR-ABL1 kinase inhibitor STI571. To investigate whether AID introduces point mutations within the BCR-ABL1 kinase domain that confer resistance to STI571, we transduced eight AID-negative CML lines with retroviral expression constructs either encoding AID/GFP or GFP alone. Within 16 days of treatment with gradually increasing concentrations of STI571, all eight AID/GFP-transduced CML lines but only one GFP-transduced CML line acquired drug resistance. Single drug resistant cells were sorted and sequence analysis confirmed that clinically relevant mutations E255K and T315I were acquired in all cases. In one cell line (K562), drug resistance and mutations were pre-existing and were not introduced by AID. Given that transcriptional activation of AID requires the B cell lineage-specific transcription factor PAX5, we transduced five CML cell lines with retroviral expression vectors encoding either PAX5/GFP or GFP alone and incubated them with STI571 at gradually rising concentrations. Outgrowth of drug-resistant subclones was observed in one of five cell lines. This outgrowth was accompanied with lineage conversion of the drug-resistant cells including cell surface expression of CD19 and upregulation of PAX5-target genes BLNK, CD79A and AID. Subsequent sequence analysis of the BCR-ABL1 kinase domain confirmed that PAX5-mediated lineage conversion has led to the acquisition of relevant BCR-ABL1 kinase mutations, likely owing to PAX5-mediated de novo expression of the mutator enzyme AID. In the four other CML cell lines, PAX5-target genes BLNK, CD79A and AID were also upregulated. However, ectopic expression of PAX5 in these cell lines did not result in CD19 surface expression and no relevant mutations were found in the BCR-ABL1 kinase domain. These findings suggest that full lineage conversion is needed to induce a, presumably AID-dependent, mutator phenotype in CML cells. We propose that B cell lineage conversion in lymphoid blast crisis CML including aberrant expression of AID accelerates the clonal evolution and induces Darwinian selection of drug-resistant mutants in CML.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 571-571
Author(s):  
Lars Klemm ◽  
Cihangir Duy ◽  
Niklas Feldhahn ◽  
John Groffen ◽  
Yong-mi Kim ◽  
...  

Abstract Chronic myeloid leukemia (CML) in chronic phase has a disease-free survival of 87% (DFS; 5 years) and long-term treatment with Imatinib is effective. In a large subgroup of patients with CML, however, the disease ultimately progresses into B lymphoid blast crisis (LBC) with only 6% DFS and resistance to Imatinib develops in virtually all cases. In most cases, acquired resistance to Imatinib can be attributed to somatic mutations within the BCR-ABL1 kinase domain. Whereas BCR-ABL1 kinase mutations are rare in chronic phase CML, such mutations are found in >80% of patients with B cell lineage LBC. Likewise, deletions of the ARF and INK4B genes are rare in chronic phase CML but found in ~50% of B cell lineage LBC. In a search for a B cell lineage-specific mutation mechanism responsible for BCR-ABL1 kinase mutations, we tested the hypothesis that aberrant activation of somatic hypermutation may give rise to drug-resistance and progression of chronic phase CML into LBC. Somatic hypermutation drives affinity maturation of immunoglobulins expressed by germinal center B cells and requires the cytidine deaminase AID. Expression of AID depends on PAX5, a transcription factor that determines B cell lineage commitment of hematopoietic progenitor cells. The dependence of AID expression on PAX5 limits somatic hypermutation to the B cell lineage. Consistent with aberrant activation of somatic hypermutation in B cell lineage LBC, we found both PAX5 and AID expression at the mRNA and protein level in B lymphoid but not myeloid subclones from patient-derived blast crisis CML. However, AID protein levels in LBC clones were 5–10-fold lower than in germinal center B cells. To confirm lineage-specific activation of AID-expression in BCR-ABL1 driven leukemia, we isolated bone marrow from Aid-GFP reporter transgenic mice and transformed the bone marrow cells with BCR-ABL1 under either myeloid (IL3, IL6, SCF) or B lymphoid (IL7) culture conditions. The Aid-GFP reporter drives GFP expression under control of upstream and downstream regulatory elements of the Aid locus (Crouch et al., 2007). BCR-ABL1-induced Aid-expression was only observed under B lymphoid culture conditions and was very heterogeneous among the leukemia cell population: Only about 5–10% of CD19+ B lymphoid leukemia clones express Aid-GFP. In these cells, however, Aid mRNA levels are 240-fold higher than in Aid-GFP-negative cells and even 1.5-fold higher than in normal germinal center B cells. Consistent with these findings, we found aberrant somatic hypermutation of the IGHM, BCL6 and MYC loci as well as evidence of ongoing DNA single-strand breaks at the ARF and INK4B loci in B cell lineage LBC but not myeloid CML clones. Ectopic expression of AID in seven otherwise AID-negative CML cell lines cells leads to the acquisition of Imatinib-resistance and sequence analysis of the Imatinib-resistant clones revealed accumulation of mutations within the BCR-ABL1 kinase domain that cause Imatinib-resistance in patients (e.g. L248V, E225K, T315I). Aberrant expression of AID also caused Imatinib-resistance of CML cells in vivo: NOD/SCID mice were injected with CML cells that were either transduced with AID/GFP or GFP alone. Whereas more than the half of the mice injected with GFP+ CML cells were still alive after 170 days, all mice in the AID/GFP+ CML group died within 54 days after injection despite Imatinib-treatment. Forced expression of the B cell-specific transcription factor PAX5 in otherwise PAX5-negative CML cells resulted in a partial B lymphoid lineage conversion similar to LBC. Of note, ectopic expression of PAX5 also resulted in aberrant AID expression, subsequent acquisition of BCR-ABL1 kinase mutations and development of drug-resistance. We conclude that B cell-specific activation of PAX5/AID-induced aberrant somatic hypermutation provides a genetic basis for the strikingly different outcome of myeloid lineage CML as compared to LBC.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 34-34
Author(s):  
Lai N Chan ◽  
Huimin Geng ◽  
Markus Muschen

Abstract Abstract 34 Background: The serine-threonine liver kinase B1 (LKB1, also called STK11) acts as negative regulator of aerobic glycoloysis, a metabolic pathway that is typically used in cancer cells (commonly referred to as ‘Warburg effect’). LKB1, together with AMPKs, shifts the metabolic balance from aerobic glycolysis to oxidative phosphorylation and thereby reverses the metabolic program of cancer cells and functions as tumor suppressor. Recently, it has been shown that LKB1 plays a critical role in the maintenance of quiescence and metabolic homeostasis of hematopoietic stem cells (HSCs). Hypothesis: In the present study, we focused on the roles of LKB1 in BCR-ABL1-driven leukemias including CML and B lymphoid blast crisis/Ph+ ALL (LBC). While LKB1 is widely seen as tumor suppressor in solid tumors, we found that high expression levels of LKB1 at diagnosis predict poor clinical outcome in patients with high risk acute lymphoblastic leukemia (n=207; COG P9906 trial; p=0.0204). In addition, high levels of LKB1 expression correlate with positive minimal residual disease (MRD+, p=0.0323) status in patients. These findings were unexpected and seem to contradict the common notion of LKB1 as a tumor suppressor. Results: To study the function of LKB1 in CML and B lymphoid blast crisis/Ph+ ALL (LBC), we developed a mouse model for inducible ablation of Lkb1 in BCR-ABL1-transformed hematopoietic stem and progenitor cells (CML-like) and B cell progenitors (LBC). To this end, Lkb1-fl/fl bone marrow hematopoietic stem and progenitor cells and B cell precursor cells were transformed with BCR-ABL1 and transduced with tamoxifen-inducible Cre. Unexpectedly, Cre-mediated deletion of Lkb1 had opposite effects in CML and LBC. While Lkb1-deletion in CML results in an initial proliferative burst of the leukemia cells, the vast majority of B cell lineage LBC cells undergo rapid cell cycle arrest. These findings are consistent with changes of cell cycle checkpoint proteins in response to Lkb1 deletion in CML and B cell lineage LBC. While Lkb1 deletion in CML cells results in downregulation of Arf, p53 and p27 levels, Lkb1 deletion in B lineage LBC cells resulted in upregulation of Arf and p27. In addition, Lkb1 deletion in CML resulted in inactivation of AMPK, a known substrate of LKB1, as well as enhanced activation of mTORC1. By contrast, while deletion of Lkb1 in B cell lineage LBC cells resulted in inactivation of AMPK as shown by reduced phosphorylation of AMPKα T172, there was reduction in mTORC1 activity based on diminished levels of phospho-p70 S6 kinase and S6 following LKB1 deletion. The effects of LKB1 on sensitivity of BCR-ABL1 CML and B lineage LBC cells to Imatinib were also examined. Lkb1-deficient ALL cells became more sensitive to Imatinib treatment. On the other hand, initial Lkb1 deletion rendered CML cells more resistant to Imatinib treatment. When primary patient-derived Ph+ ALL cells (n = 3) were treated with Imatinib, upregulation of phospho-LKB1 (S428) was observed. Finally, LKB1 was also shown to regulate energy homeostasis in CML and B cell lineage LBC in different manners, as measured by monitoring ATP and lactate production. Conclusions: Here we show that Lkb1 plays critical roles in mediating proliferation and cell growth in BCR-ABL1-driven leukemias. While LKB1 is widely seen as a tumor suppressor that limits aerobic glycolysis in cancer cells according to the Warburg effect, our findings demonstrate that LKB1 has lineage-specific functions in BCR-ABL1 driven leukemias. While LKB1 function in CML resembles its tumor suppressor function in solid tumors, LKB1 is critical for survival and proliferation on B cell lineage CML blast crisis and Ph+ ALL. The finding of a divergent role of Lkb1 in CML and B cell lineage LBC/Ph+ ALL is relevant because small molecule inhibitors of AMPK and mTORC1 are currently under development for the treatment of BCR-ABL1-driven leukemias. Disclosures: No relevant conflicts of interest to declare.


2005 ◽  
Vol 161 (2) ◽  
pp. 187-188
Author(s):  
Bonaventure Ndikung Bejeng Soh ◽  
Florian Klein ◽  
Niklas Feldhahn ◽  
Markus Müschen

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3274-3274
Author(s):  
Lars Klemm ◽  
Cihangir Duy ◽  
Ilaria Iacobucci ◽  
Gregor von Levetzow ◽  
Niklas Feldhahn ◽  
...  

Abstract Abstract 3274 Poster Board III-1 Chronic myeloid leukemia (CML) is induced by BCR-ABL1 and can be effectively treated for many years with Imatinib until leukemia cells acquire drug resistance through BCR-ABL1 mutations and progress into fatal B lymphoid blast crisis (LBC). Treatment of patients with CML in chronic phase with the BCR-ABL1 kinase inhibitor Imatinib leads to a five-year overall survival of ∼95%. Upon blast crisis progression, however, median overall survival decreases to less than seven months and CML blasts are frequently Imatinib-resistant due to BCR-ABL1 mutations. Despite its clinical significance, the mechanism of progression into LBC is unknown. Here we show that LBC but not CML cells aberrantly express the B cell-specific mutator enzyme AID. During normal B cell development, AID is activated within germinal centers to initiate somatic hypermutation and class-switch recombination of immunoglobulin genes during affinity maturation of B cells. When ectopically expressed in CML cells, AID promotes widespread hypermutation and thereby promotes B lymphoid blast crisis progression. To examine whether aberrant mutational activity of Imatinib leads to Imatinib-resistance in vivo, we then labeled GFP+> and AID/GFP+> transduced CML cells with lentiviral firefly luciferase and injected them intrafemorally into sublethally irradiated NOD/SCID mice. When focal expansion of leukemic growth became evident (day 20), treatment with Imatinib (100 mg/kg twice daily) was started. By day 35, all 14 mice injected with AID/GFP-transduced CML cells had developed progressive disease as opposed to only 5 of 12 GFP-transduced control mice (P <0.02). Correspondingly, all mice injected with AID/GFP+> CML cells were dead at day 54, whereas about half of the mice injected with GFP+> CML cells were still alive 162 days post-injection (n=12; P<0.001).In a genetic loss-of-function experiment, we tested the ability of AID+/+> and AID-/-> BCR-ABL1-transformed leukemia cells to acquire Imatinib-resistance and BCR-ABL1 mutation. AID+/+> but not AID-/-> BCR-ABL1-transformed leukemia cells became Imatinib-resistant within six under cell culture conditions. Sequence analysis revealed that most of the AID+/+> Imatinib-resistant clones had acquired in the BCR-ABL1 kinase domain that confer Imatinib-resistance in patients. From AID-/-> leukemia cells, only one clinically relevant mutation was amplified. To determine whether AID promotes overall genetic instability in BCR-ABL1 leukemias, we measured gene copy number alterations (i.e. deletions or amplifications) in 23 primary cases of Ph+> ALL using a 250K NspI SNP array. Based on AID mRNA levels, Ph+> ALL samples were classified as either AIDhigh> (16 cases) or AIDlow> (7 cases). SNP analysis revealed a higher frequency of gene copy number alterations in the AIDhigh> as compared to the AIDlow> group (median 14 [range 6–50] vs. median 5 [range 2–8]; P = 0.02). Notably, deletion frequencies at the tumor suppressor genes ARF (CDKN2A) and INK4B (CDKN2B) at 9p21 were considerably higher in the presence of AID (P = 0.04). The transcription factor PAX5 regulates both AID-gene expression and B cell lineage commitment of hematopoietic progenitor cells. Interestingly, ectopic expression of PAX5 in myeloid CML cells was sufficient to induce AID transcription and B cell lineage conversion including expression of CD19 in a small subset of PAX5/GFP+> but not in GFP+> CML cells. To determine whether PAX5 can promote Imatinib-resistance in CML, PAX5/GFP+> and GFP+ >human leukemia cells were cultured in the presence of increasing concentrations of Imatinib (0.1 μmol/L to 1.75 μmol/L). A time-dependent outgrowth of PAX5/GFP+ >CML cells in the presence of Imatinib was observed and sequence analysis confirmed the presence of mutations within the BCR-ABL1 kinase domain of PAX5/GFP but not in GFP-transduced CML cells. We conclude that enforced PAX5 expression in myeloid CML cells can lead to partial B cell lineage conversion, AID expression, and Imatinib-resistance via BCR-ABL1 mutation. We demonstrate that AID expression in CML cells promotes overall genetic instability by hypermutation of tumor suppressor and DNA repair genes. Importantly, our data uncover a causative role of AID activity in the acquisition of BCR-ABL1 mutations leading to Imatinib-resistance, thus providing a rationale for the rapid development of drug resistance and blast crisis progression. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3128-3128
Author(s):  
Gang Xiao ◽  
Huimin Geng ◽  
Lai N Chan ◽  
Zhengshan Chen ◽  
Markus Muschen

Abstract Background & Hypothesis: Protein phosphatase 2A (PP2A) is a Ser/Thr phosphatase and functions as negative regulator of Akt- and Erk-signaling. PP2A attenuates downstream signaling of BCR-ABL1 and thereby functions as important tumor suppressor in chronic myeloid leukemia (CML). PP2A-activating drugs have been proposed for the treatment of CML and eradication of tyrosine kinase inhibitor resistant leukemia stem cells in CML (Neviani et al., 2013). Studying Cre-mediated ablation of PP2A in mouse models for BCR-ABL1-driven leukemia including myeloid CML in chronic phase (CML-CP), B lymphoid blast crisis (LBC) and B cell lineage Ph+ ALL, we were able to provide genetic evidence for this treatment concept in CML. Seemingly paradoxically, however, inducible ablation of PP2A in B cell lineage LBC and Ph+ ALL cells has the opposite effect and induced rapid cell death. These findings highlight fundamental differences in the regulation of Akt- and Erk-signaling in myeloid lineage and B cell lineage cells and have implications for specific therapy concepts that distinguish between chronic phase CML and lymphoid blast crisis/ Ph+ ALL. Results: Interestingly, PP2A protein levels are significantly higher in B cell lineage Ph+ ALL and CML lymphoid blast crisis (LBC) compared to chronic phase CML. To explore the function of PP2A during early B cell development, we deleted the α isoform of the PP2A subunit A in pro-B cells by introducing Mb1-Cre into Ppp2r1afl/fl mice. Analysis of bone marrow from Ppp2r1afl/flMb1-Cre (PP2A-cko) mice showed obvious B cell development block at the pre-B cell receptor checkpoint, although V(D) recombination and RAG1/2 activity were intact in PP2A-cko pre-B cells. Since high mRNA levels of PP2A subunits at the time of diagnosis predict poor outcome of children (COG P9906; n=207) and adults (ECOG 2993; n=215) with ALL, we studied the function of PP2A in mouse models for B cell lineage ALL including Ph+ ALL and CML lymphoid blast crisis (LBC). After Cre-induced deletion of Ppp2r1a, we observed reduced protein expression of both PP2A subunit A and subunit C accompanied with decreased PP2A phosphatase. The PP2A-cKO ALL cells showed normal proliferation but significantly impaired colony formation capability and increased apoptosis, both of which could be rescued by overexpression of wildtype PP2A in PP2A-cKO ALL cells. We next transplanted luciferase expressing PP2A-KO ALL cells into recipient mice and monitored cell growth and leukemia progression. Cre-mediated deletion significantly prolonged overall survival of recipient mice that were transplanted with Ppp2r1afl/fl ALL cells. Although all of those mice died eventually from leukemia, the ALL cells isolated from the mice retained Ppp2r1a floxed alleles suggesting that these mice died from clones that had escaped Cre-mediated deletion of PP2A. Cre-mediated deletion increased phosphorylation levels of Stat5 and Erk, resulted in accumulation of p53 and increased output of the PI3K-AKT signaling pathway as measured by increased phosphorylation of FoxO factors, p70S6K and S6 ribosomal protein. Based on the involvement of PI3K-AKT in glucose metabolism, we measured the effects of inducible PP2A-deletion on glycolytic function in CML and Ph+ ALL/LBC cells. Interestingly, inducible deletion of PP2A induced profound imbalances of glucose metabolism in B cell lineage Ph+ ALL/LBC but not myeloid lineage CML-like cells. Upon PP2A-deletion, ALL cells showed higher glycolytic flux shunted into lactate rather than NADPH production. Lower NADPH/NADP ratio and higher reactive oxygen species level in PP2A-KO ALL cells indicated impaired balance of glycolytic flux and may account for increased apoptosis of those cells. This notion was supported by a strong rescue effect of overexpression of the antioxidant catalase in PP2A-cKO cells. In agreement with reported tumor suppressor role of PP2A in CML cells (Neviani et al., 2005), we observed that CEBPα-driven lineage conversion of B cell lineage Ph+ ALL/LBC into not myeloid lineage. Conclusion: Here we reported the requirement of PP2A in normal B cell development and lineage-specific oncogenic role of PP2A in BCR-ABL1-driven leukemia. Divergent roles of PP2A in Ph+ ALL/LBC and myeloid lineage CML may lead to future insights into mechanisms of LBC-transformation of CML and lineage-specific requirement of BCR-ABL1-transformation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1436-1436
Author(s):  
Tomasz Sewastianik ◽  
Patryk Gorniak ◽  
Przemyslaw Kiliszek ◽  
Anna Polak ◽  
Emilia Bialopiotrowicz ◽  
...  

Abstract Precursor B-cell lymphoblastic leukemia (B-ALL) in adults remains a challenging clinical problem due to higher relapse rate and worse prognosis than in children. Although most of adult patients respond to standard induction therapy and complete remissions (CR) are typically achieved in 90% of patients, the majority of them eventually relapse. Our previous studies indicate that the minimal residual disease (MRD) status after induction therapy is the most important risk factor of relapse in adult B-ALL patients [Br J Haematol 2008;142:227-37]. We hypothesized that the survival of B-ALL blasts after induction therapy is a result of intrinsic characteristics of the tumor cells that determine resistance to chemotherapeutics. Therefore, we sought to identify the molecular background of B-ALL cells resistance to daunorubicin. To identify potential mechanisms responsible for drug-resistant phenotype, we utilized gene expression data from previous studies that assessed transcriptional profiles of drug-sensitive and drug-resistant cells. Using gene set enrichment analysis (GSEA) of daunorubicin-sensitive versus -resistant phenotypes of B-ALL cells we identified differential expression HIF1α and MYC transcription factors target genes (p=0.002, FDR=0.144; p<0.001; FDR=0.171, respectively). To verify these in silico findings, we compared the expression of a panel of MYC and HIF1α target genes in 41 newly diagnosed adult B-ALL patients who subsequently underwent standard induction therapy according to Polish Adult Leukemia Group PALG-ALL6 protocol. Expression of MYC and HIF1α signature genes was significantly higher in patients with positive (>0.1%) MRD status after completion of the induction therapy. Among studied HIF1α and MYC targets, lactate dehydrogenase A (LDHA) expression was the best predictor differentiating MRD+ versus MRD- patients (p=0.0019, FDR=0.005). It was of particular interest, since tumor stem cells are typically characterized by MYC and HIF1α transcriptional signatures, which rewire cellular metabolism towards aerobic glycolysis. We next assessed the effect of LDHA inhibition with a small molecule inhibitor, GSK2837808A, on proliferation and clonogenicity of human B-ALL cell lines. GSK2837808A markedly reduced lactate production in B-ALL cell lines (RS4;11, SEM-K2 and NALM-6) and decreased proliferation and colony formation in semi-solid medium in a dose-dependent fashion. Taken together, we show that adult B-ALL patients with positive MRD status after induction therapy exhibit concordant upregulation of HIF1α and MYC signature genes. Expression of LDHA, a target gene regulated by both HIF1α and MYC transcription factors was significantly higher in MRD-positive patients. Finally, inhibition of LDHA markedly decreased proliferation and clonogenicity of B-ALL cell lines, indicating that LDHA might be a therapeutic target in B-ALL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 902-902
Author(s):  
Xiao Gang ◽  
Huimin Geng ◽  
Lai N Chan ◽  
Zhengshan Chen ◽  
Xiaoyan Jiang ◽  
...  

Abstract Background: The Ser/Thr Protein phosphatase 2A (PP2A) attenuates activity of RAS-ERK and PI3K-AKT signaling pathways and functions as important tumor suppressor in chronic myeloid leukemia (CML) by downregulating output of oncogenic BCR-ABL1 signaling. Restoration of PP2A activity has been proposed for the treatment of CML and eradication of TKI-resistant leukemia CML-stem cells (Neviani et al., 2013). While the tumor suppressor function of PP2A was independently confirmed by multiple groups, our analyses of PP2A function during early B cell development suggested a role for PP2A to contribute to B cell survival. Therefore, we studied whether PP2A has different roles in B-lineage ALL and CML and whether these mechanistic differences are relevant for patients with B-lineage and B-lymphoid blast crisis CML. Results: Consistent with a divergent role of PP2A in B-lineage ALL (compared to CML), we found that high mRNA levels of PP2A subunits at the time of diagnosis predict poor outcome of children (COG P9906; n=207) and adults (ECOG 2993; n=215) with ALL. We therefore, studied the function of PP2A in a genetic mouse model for Cre-induced deletion of Ppp2r1a in BCR-ABL1 (Ph+) ALL. Inducible activation of Cre reduced protein expression of the targeted PP2A subunit A but also the catalytic subunit C and was paralleled by near-complete loss of PP2A phosphatase activity. Conversely, Cre-mediated deletion increased phosphorylation levels of p70S6K and S6 ribosomal protein, which indicated elevated mTOR signaling. Acute deletion of Ppp2r1afl/fl in B cell-lineage ALL cells dramatically affected survival and colony formation, both of which could be rescued by overexpression of wildtype PP2A. In agreement with previous studies, however, Cre-mediated deletion had no deleterious effects in a Ppp2r1afl/fl CML model. To verify that the observed difference between B cell and myeloid disease was indeed lineage-dependent, we reprogrammed B cell lineage ALL cells into myeloid CML using inducible overexpression of CEBPα. As expected, CEBPα-driven myeloid lineage conversion prevented cell death upon deletion of PP2A, demonstrating that PP2A-function represents a B cell-specific vulnerability. We next transplanted luciferase-labeled Ppp2r1afl/fl ALL cells into recipient mice and monitored cell growth and leukemia progression. Cre-mediated deletion significantly prolonged overall survival of recipient mice that were transplanted with Ppp2r1afl/fl ALL cells. Although mice died eventually from leukemia, the ALL cells isolated from the mice retained Ppp2r1a floxed alleles in all cases studied demonstrating that fatal leukemia arose from few subclones that had escaped Cre-mediated deletion of PP2A. Interestingly, inducible deletion of PP2A caused profound imbalances of glucose metabolism in B cell lineage Ph+ ALL but not CML-like cells. Upon PP2A-deletion, ALL cells showed higher glycolytic flux shunted into lactate rather than NADPH production. Lower NADPH/NADP ratio and higher reactive oxygen species level in PP2A-deleted ALL cells, together with decreased anti-oxidant gene expression, increased H2AX phosphorylation and p53 expression indicated impaired balance of glycolytic flux may account for increased death of those cells. This notion was supported by a strong rescue effect of overexpression of the antioxidant catalase in PP2A-deleted cells. The unexpected role of PP2A in Ph+ ALL was further validated by CRISPR-Cas9 mediated disruption of PPP2R1A in ALL xenografts derived from three patients. In addition, a PP2A specific inhibitor LB-100 (in clinical trial for solid tumors) was employed to pharmacologically inhibit PP2A activity. Low micromolar concentrations of LB-100 induced cell death in patient-derived ALL xenografts in parallel with ROS-accumulation and increased S6 and H2AX phosphorylation. Conclusion: Cre-mediated ablation of PP2A in mouse Ph+ ALL cells induced rapid cell death through excessively oxidative stress but not in myeloid CML cells. We confirmed this pro-survival role of PP2A in human Ph+ ALL-patients derived leukemia cells through both CRISPR-Cas9 mediated genetic study and PP2A inhibition by a novel small molecule LB-100 (Jie Lu et al., 2009). These findings highlight PP2A as a therapeutic target with potential relevance in Ph+ ALL and B-lymphoid blast crisis progression of CML. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 513-513
Author(s):  
Eugene Park ◽  
Srividya Swaminathan ◽  
Mohammed Firas Sadiyah ◽  
Kazuhiko Igarashi ◽  
Ari Melnick ◽  
...  

Abstract Background and Hypothesis: CML and Ph+ ALL are both driven by the oncogenic BCR-ABL1 tyrosine kinase and CML can progress into lymphoid blast crisis (LBC), which is clinically and biologically indistinguishable from Ph+ ALL. We hypothesize that the stark differences in phenotype and clinical outcomes between CML and Ph+ ALL/LBC are due to lineage-specific transcription factors. Our group recently identified BACH2 as a B cell-specific transcription factor that mediates negative selection at the pre-B cell receptor checkpoint and functions as a tumor suppressor in Ph+ ALL and LBC (Swaminathan et al., Nature Med 2013). Here we report the surprising finding that BACH2 mediates lineage-specific fate decisions in BCR-ABL1 driven leukemia: while a potent tumor suppressor in B cell lineage in Ph+ ALL and LBC, BACH2 is required for survival and self-renewal of myeloid lineage CML cells. Results: A gene expression analysis in 99 patients with CML (Radich et al, 2006) revealed that patients with higher blast counts showed significantly higher gene expression values of BACH2 (P=8.88 x 10-9), suggesting that BACH2 mRNA levels increase with CML progression. To determine the mechanistic role of BACH2 in the progression of CML, we studied genetic deletion of BACH2 in a CML mouse model. To this end, we transformed Lin-kit+Sca-1+(LSK) cells from Bach2+/+ and Bach2-/- bone marrow with BCR-ABL1. We compared these cells alongside with Bach2+/+ and Bach2-/- pre-B cells that were transformed with BCR-ABL1 as a model for Ph+ ALL and LBC. Bach2+/+ and Bach2-/- CML-like and Ph+ ALL/LBC-like leukemia cells were then tested in a series of functional experiments to test their ability to initiate fatal leukemia in transplant recipients. While deletion of BACH2 accelerated B cell lineage leukemia (Ph+ ALL/LBC-like) in transplant recipients, we noted the opposite outcome in transplant experiments with myeloid lineage CML cells: All mice receiving Bach2+/+BCR-ABL1-transformed LSK cells developed CML-like disease within 60 days whereas recipients of Bach2-/- cells did not develop CML-like disease (Bach2+/+ vs Bach2-/-, median survival time= 33 days vs undefined; P=0.001). Additionally, we observed that deletion of Bach2 loss reduced both the colony formation ability (P=0.0059) and the S phase proliferation potential (P=0.0075) of the CML progenitors. These results were in contrast to those observed in Ph+Bach2+/+ and Bach2-/- ALLs. Reconstitution of Bach2 in Bach2-/- CML cells rescued its colony forming ability (P= 0.023). These findings were confirmed in primary human CML cells from two patients with CML in chronic phase: Inducible overexpression of Bach2 in patient-derived CML cells conferred a selective proliferative advantage of BACH2 overexpressing cells over time, in comparison to empty vector controls. Bach2 triggered a survival program in human and mouse CML cells, in contrast to B cell lineage Ph+ ALL and LBC cells. Interestingly, Bach2 did not provide a selective growth advantage to untransformed myeloid and multi-lineage progenitor cells. This demonstrates that Bach2 drives lineage-specific fate decisions and cooperates with the transforming BCR-ABL1 oncogene. Utilizing an in vitro lineage-switch assay overexpressing CEBPα we characterized the opposing roles of BACH2 in Ph+ ALL and CML. Cell viability and colony formation assays demonstrated that absence of Bach2 results in increased survival and proliferation of Ph+ ALL cells. In striking contrast, however, CEBPα-induced myeloid reprogramming induced cell cycle arrest and suppressed colony formation in Bach2-deficient cells (P=0.0001). Conclusion: While BACH2 is primarily expressed in the B cell lineage, functioning as a tumor suppressor in Ph+ ALL, we report here the surprising finding that BACH2 is required for survival and self-renewal of myeloid lineage CML cells. BACH2 mediates lineage-specific fate decisions in BCR-ABL1 driven leukemia and represents one example of how conversion of lineage identity (e.g. from CML to LBC) may impact specific requirement for malignant transformation. For instance, deletions of BACH2 at 6q15 are common in CML at the time of LBC progression but are not found in CML chronic phase or myeloid blast crisis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1985 ◽  
Vol 66 (2) ◽  
pp. 342-344 ◽  
Author(s):  
AM Ferraris ◽  
WH Raskind ◽  
BH Bjornson ◽  
RJ Jacobson ◽  
JW Singer ◽  
...  

Abstract In order to study the pattern of B cell involvement in acute nonlymphocytic leukemia (ANLL), multiple B lymphoid cell lines were established by Epstein-Barr virus transformation of peripheral blood mononuclear cells from two patients with the disease who were heterozygous for the X chromosome-linked glucose-6-phosphate dehydrogenase (G6PD). In one patient, the progenitor cells involved by the leukemia exhibited multipotent differentiative expression, whereas in the other patient the cells showed differentiative expression restricted to the granulocytic pathway. In the patient whose abnormal clone showed multipotent expression, the ratio of B-A G6PD in B lymphoid cell lines was skewed in the direction of type B (the enzyme characteristic of the leukemia clone) and significantly different from the 1:1 ratio expected. It is, therefore, likely that the neoplastic event occurred in a stem cell common to the lymphoid series as well as to the myeloid series. In contrast, evidence for B cell involvement was not detected in the patient whose ANLL progenitor cells exhibited restricted differentiative expression. These findings underscore the heterogeneity of ANLL. Clinically and morphologically similar malignancies in these two patients originated in progenitors with different patterns of stem cell differentiative expression. This difference may reflect differences in cause and pathogenesis.


Sign in / Sign up

Export Citation Format

Share Document