Summary
Reprogrammed cellular metabolism is a common characteristic observed in various cancers1,2. However, whether metabolic changes directly regulate cancer development and progression remains poorly understood. Here we show that BCAT1, a cytosolic aminotransferase for the branched-chain amino acids (BCAAs), is aberrantly activated and functionally required for chronic myeloid leukemia (CML). BCAT1 is up-regulated during CML progression and promotes BCAA production in leukemia cells by aminating the branched-chain keto acids. Blocking BCAT1 expression or enzymatic activity induces cellular differentiation and impairs the propagation of blast crisis CML (BC-CML) both in vitro and in vivo. Stable isotope tracer experiments combined with NMR-based metabolic analysis demonstrate the intracellular production of BCAAs by BCAT1. Direct supplementation with BCAAs ameliorates the defects caused by BCAT1 knockdown, indicating that BCAT1 exerts its oncogenic function via BCAA production in BC-CML cells. Importantly, BCAT1 expression not only is activated in human BC-CML and de novo acute myeloid leukemia but also predicts disease outcome in patients. As an upstream regulator of BCAT1 expression, we identified Musashi2 (MSI2), an oncogenic RNA binding protein that is required for BC-CML. MSI2 is physically associated with the BCAT1 transcript and positively regulates its protein expression in leukemia. Taken together, this work reveals that altered BCAA metabolism activated through the MSI2-BCAT1 axis drives cancer progression in myeloid leukemia.
To understand the contribution of α-amino acid (AA) metabolism to the cancer progression of CML, we analyzed blood AA levels in murine models that recapitulate the chronic and blast crisis phases of human CML3,4. Using amine-specific fluorescent labeling coupled with high-performance liquid chromatography, sixteen AAs were quantified in the blood plasma from leukemic mice (Extended Data Fig. 1a–d). Mice bearing BC-CML showed moderate but significant elevations of plasma glutamate, alanine and the branched-chain amino acids (BCAAs; namely, valine, leucine and isoleucine) compared to CP-CML mice, indicating hyperaminoacidemia (Extended Data Fig. 1e). Intracellular levels of BCAAs and proline were higher in BC-CML, whereas intracellular glutamate and alanine were comparable in the two disease phases (Fig. 1a). These results suggest that increased BCAA uptake or metabolism may contribute to CML progression. We analyzed the gene expression and found no significant up-regulation of known BCAA transporters in BC-CML compared with CP-CML (data not shown). Leucine import into BC-CML cells was not greater than into CP-CML cells (Extended Data Fig. 1f), indicating that increased BCAA uptake does not explain the higher BCAA levels in BC-CML. To examine the possibility of altered intracellular BCAA metabolism, we next analyzed the expression of genes encoding AA metabolic enzymes and found that the branched-chain amino acid aminotransferase 1 (Bcat1) was more highly expressed in BC-CML than in CP-CML at both the mRNA and protein levels (Fig. 1b–c, Extended Data Fig. 1g–h). In contrast, normal hematopoietic stem/progenitor cells (HSPCs) from healthy mice had very low levels of Bcat1 expression (Lin− Sca-1+ cKit+ (LSK) population; Fig. 1b), and normal tissues did not show detectable Bcat1 expression except for the brain and testis (Extended Data Fig. 1i). Bcat1 encodes an evolutionarily conserved cytoplasmic aminotransferase for glutamate and BCAAs, constituting a regulatory component of cytoplasmic amino and keto acid metabolism5 (Fig. 1d). Bcat2, a paralog encoding the mitochondrial BCAA aminotransferase, and alanine and aspartate aminotransferases did not show differential expression between CP- and BC-CML (Extended Data Fig. 1g–l).
Although BCAT1 catalyzes transamination in both directions, the breakdown of BCAAs is the predominant reaction in most cell types6. In order for BCAT1 to generate BCAAs via the reverse reaction, the corresponding branched-chain keto acids (BCKAs), as well as glutamate, must be present as substrates. We found all three BCKAs, keto-isovalerate (KIV), keto-isocaproate (KIC) and keto-methylvalerate (KMV), were present in both the blood plasma and leukemia cells (Extended Data Fig. 2a–d). In BC-CML cells, BCKAs were present at concentrations equivalent to 22–55% of the corresponding BCAAs, suggesting that intracellular BCKAs can serve as substrates for BCAA production (Extended Data Fig. 2e). Next, we examined whether BCAAs are produced through BCAT1 transamination reactions in leukemia cells by stable-isotope tracer experiments with 13C-valine or 13C-KIV. Intracellular 13C-labeled metabolites in K562 human BC-CML cells were analyzed using one- (1D) and two-dimensional (2D) 1H-13C heteronuclear single bond correlation (HSQC) analysis by high-field NMR spectroscopy (Fig. 1e–h, Extended Data Fig. 3). HSQC analysis detects only metabolites that have incorporated 13C. To determine whether KIV is converted to valine, cells were cultured in media supplemented with uniformly-labeled [(U)-13C] KIV and non-labeled valine at physiological concentrations (30 and 170μM, respectively) and analyzed for intracellular 13C-metabolites. After 15 min of labeling, the generation of 13C-valine was clearly observed, indicating the efficient intracellular production of valine from KIV (Fig. 1f, h). In contrast, 13C-KIV formation was barely detectable in the cells cultured with non-labeled KIV and [(U)-13C]-valine (Fig. 1e, g). Our observation of intracellular 13C-valine signals indicates its transport into BC-CML cells. We also detected robust signals for 13C-KIV when present (Extended Data Fig. 3d, f). The formation of valine from KIV, but not the breakdown of valine to KIV, was also observed when we used equal concentrations of KIV and Val in the labeling media (170μM each; Fig.1g–h). We did not detect KIC formation from 13C-leucine either (Extended Data Fig. 3g–i). These results indicate that little, if any, BCAAs are catabolized to BCKAs in leukemia cells. To further provide evidence for the intracellular BCAA production through transamination, we performed alternative labeling experiments to track the fate of the amine group of glutamate. We cultured K562 cells with 15N-amine-labeled glutamine, which is metabolized to 15N-amine-glutamate by glutaminase upon cellular intake, and analyzed the subsequent labeling of BCAAs via 1H NMR and 1H-15N heteronuclear multiple bond correlation (HMBC) analysis. HMBC analysis detects only metabolites that have incorporated 15N, whereas 1H NMR detects any compounds containing protons (Extended Data Fig. 4a–f). At 29–72 h post-labeling, we detected 15N-amine-labeled BCAAs, indicating transamination from glutamine to BCAAs (Fig. 1i). By 72 h, the 15N-amine-labeled BCAAs had accumulated to fractional abundances ranging from 24 to 39% (Extended Data Fig. 4g), indicating a significant contribution of transamination to the intracellular BCAA pool. Lentiviral BCAT1 knockdown resulted in greater than a 50% decrease in the amount of intracellular BCAAs produced (Fig. 1j). These data demonstrate that BCKA transamination by BCAT1 contributes to the BCAA pool in leukemia cells.
Given that Bcat1 is highly expressed and augments intracellular BCAAs in BC-CML, Bcat1 may functionally contribute to the acute properties of BC-CML. To test this possibility, we inhibited Bcat1 expression using a short hairpin RNA (shRNA)-mediated gene knockdown approach. We sorted the immature lineage-negative (Lin−) cells from primary BC-CML samples, a population that contains the leukemia-initiating cells of this cancer, and introduced two independent retroviral shRNA constructs (Extended Data Fig. 1j; shBcat1-a and shBcat1-b). Both constructs inhibited Bcat1 expression in BC-CML compared with a non-targeting negative control shRNA (shCtrl) (Extended Data Fig. 5a–c). Bcat1 knockdown resulted in significantly smaller colonies and a 40–60% reduction in the colony-forming ability relative to a control (Fig. 2a). The co-introduction of a shRNA-resistant Bcat1 cDNA rescued the reduced clonogenic potential (Extended Data Fig. 5d). As an alternative approach to gene knockdown, we treated BC-CML cells with gabapentin (Gbp), a chemical inhibitor of BCAT1. Gbp is a structural analog of leucine and specifically and competitively inhibit the transaminase activity of BCAT1 but not that of BCAT27. BC-CML cells plated with Gbp formed smaller colonies and showed a dose-dependent impairment in clonogenic growth (Fig. 2b). In contrast, normal HSPCs were only minimally affected by gene knockdown or Gbp treatment (Extended Data Fig. 5e–f). These data suggest that BCAT1 inhibition may selectively impair the propagation of leukemia without affecting normal hematopoiesis.
To examine whether Bcat1 loss affects the propagation of BC-CML in vivo, Lin− cells expressing shBcat1 were transplanted into conditioned recipient mice. Whereas 75% of the recipients transplanted with control cells succumbed to the disease within 30 days, only 47% (shBcat1-a) and 31% (shBcat1-b) of the mice transplanted with Bcat1-knockdown cells developed the disease, and more than half of these mice survived even when followed out to 60 days (Fig. 2c). Among the mice that developed disease with Bcat1 knockdown, most had leukemia that was characterized by differentiated granulocytes and lower levels of immature myeloblasts (Fig. 2d, Extended Data Fig. 4g). They also displayed a lower frequency of immature Lin− cells than control leukemia (Extended Data Fig. 5h), indicating that the loss of Bcat1 induced differentiation and impaired the leukemia-initiating cell activity. Consistent with these phenotypes, serial transplantation of the leukemia cells revealed that while all the control leukemia propagated the disease, none of the mice transplanted with Bcat1-knockdown leukemia cells succumbed to the disease (1k; Fig. 2e). In addition, we established a doxycycline (Dox)-inducible Bcat1 knockdown system (i-shBcat1) and examined the impact of Bcat1 loss on the disease maintenance. Ten days post-transplantation with BC-CML cells infected with i-shBcat1, leukemic engraftment was assessed in each recipient, and Dox treatment was initiated (Extended Data Fig. 5i–j). While almost all the mice that were transplanted with control cells and the non-Dox-treated mice developed leukemia, more than half of the Dox-treated i-shBcat1 mice remained disease-free (Extended Data Fig. 5k), indicating that Bcat1 is required for the continuous propagation of BC-CML. At the cellular level, we did not observe enhanced apoptosis or a decrease in actively cycling cells by Bcat1 knockdown (Extended Data Fig. 5l–m). These results demonstrate that Bcat1 is critical for the sustained growth and maintenance of leukemia-initiating cells in BC-CML.
We next examined whether the enforced expression of Bcat1 could drive blastic transformation in hematopoietic cells. Although we observed a significant increase in Bcat1 expression compared with the vector control, Bcat1 expression alone did not enhance the colony-forming ability of either LSK or Lin− c-Kit+ hematopoietic cells isolated from normal bone marrow (Extended Data Fig. 6a–b). To determine whether BCR-ABL1 cooperates with Bcat1 overexpression to confer an aggressive growth phenotype, we transduced normal HSPCs with Bcat1 and BCR-ABL1. Compared with the vector control, the combinatorial expression promoted clonogenic growth in vitro (Extended Data Fig. 6c), and the transplantation of the cells led to significantly elevated leukemia burdens (Extended Data Fig. 6d–e), splenomegaly and increased mortality in the recipient mice (Fig. 2f), with a concomitant increase in plasma BCAA levels (Extended Data Fig. 6f). Accordingly, leukemia that developed in response to Bcat1 overexpression exhibited a highly immature myeloblastic morphology compared to the control (Fig. 2g, Extended Data Fig. 6g). These data indicate that activated Bcat1 mediates the blastic transformation of CP-CML cells.
Our results demonstrate that Bcat1 is essential for the development of BC-CML in mice, while normal bone marrow HSPCs show a very limited dependence on this metabolic enzyme. To investigate the contribution of BCAT1 to human leukemia, we looked at a panel of 13 peripheral blood samples from healthy and leukemic subjects and found human BCAT1 expression was higher in BC-CML than in either normal or CP-CML cells (Fig. 3a). To determine whether this expression pattern reflects a general trend in human CML, we analyzed BCAT1 levels in a GEO dataset of 113 CML patient samples8. This focused analysis revealed a significant elevation in BCAT1 expression as the disease progresses from the chronic to the accelerated phase and then to the blast crisis phase (Fig. 3b). On average, BCAT1 expression was 15-fold higher in BC-CML than in CP-CML. We did not find significant changes in BCAT2 expression, which is consistent with the results from the mouse models (Fig. 3c, Extended Data Fig. 1g). These data indicate that activation of BCAT1 is a shared characteristic in the progression of human CML. Lentiviral BCAT1 knockdown or Gbp treatment markedly inhibited the colony-forming ability of K562 human BC-CML (Extended Data Fig. 7a–d) and patient-derived primary leukemia cells (Fig. 3d–e, Extended Data Fig. 7e–f). Interestingly, we observed BCAT1 activation in primary human acute myeloid leukemia as well (AML; Fig. 3f), and Gbp effectively inhibited the clonal growth of human AML cell lines and primary de novo AML cells (Fig. 3g, Extended Data Fig. 7g–i). Moreover, BCAT1 expression levels predict disease outcome in patient cohorts. Cases from the TCGA AML dataset were divided into quartiles based on BCAT1 expression levels (Extended Data Fig. 7j), and we found that the median survival time was 46% shorter in the BCAT1-high group (427 vs. 792 days; Fig. 3h). These results demonstrate an essential role for BCAT1 in the pathogenesis of a wide array of human myeloid malignancies.
To understand how the BCAT1-driven change in metabolism promotes leukemia growth, we analyzed intracellular AA concentrations upon BCAT1 inhibition and found that all three BCAAs were significantly reduced by shBCAT1 or Gbp treatment compared with the controls (Extended Data Fig. 8a–b). Interestingly, the addition of BCAAs, but not alanyl-glutamine (GlutaMax), functionally suppressed the reduction of colony-forming ability caused by BCAT1 knockdown (Fig. 3i), suggesting that BCAT1 enhances clonogenic growth through BCAA production via BCKA reamination. BCAAs, particularly leucine, activate the mTORC1 pathway via cytosolic leucine sensor proteins, which integrate multiple signals from nutrient sensing and growth factor stimuli to promote cell growth9–12. Thus, we examined whether reduced BCAA production by BCAT1 inhibition results in the attenuation of the mTORC1 signal. Indeed, BCAT1 blockade by either shRNA or Gbp treatment significantly reduced the phosphorylation of S6 kinase (pS6K), a downstream target of mTORC1 kinase (Fig. 3j), suggesting BCAT1 activation of the mTORC1 pathway. We observed no apparent changes in the levels of phosphorylated AKT upon BCAT1 inhibition, suggesting a predominant contribution of BCAA nutrient signals to the activation of mTORC1 (Extended Data Fig. 8c). Consistently, the mTORC1 inhibitor rapamycin reversed the BCAA-induced suppression of colony formation (Fig. 3i) and the BCAA-induced increase in pS6K (Fig. 3k).
To further investigate the BCAT1-mediated regulation of CML progression, we performed gene correlation analyses using tumor gene expression datasets available in the GEO and TCGA databases. We found that BCAT1 and MSI2 are often co-expressed in several types of cancer, including leukemias, colorectal and breast cancers (Extended Data Fig. 9a–b). MSI2 is a member of the evolutionarily conserved Musashi RNA binding protein family, which regulates cell fates during development and in multiple adult stem cell systems in metazoans13–15. At the molecular level, Musashi proteins bind to r(G/A)U1-3AGU sequences (MSI binding elements, MBEs) and post-transcriptionally regulate gene expression via mRNA binding16,17. Importantly, MSI genes are aberrantly activated in human malignancies, such as gliomas and breast and colorectal cancers18,19. In human BC-CML, the MSI2 gene is up-regulated and functionally required for the progression of this leukemia20,21. To determine whether BCAT1 is a direct target of the MSI2 RNA binding protein, we analyzed the BCAT1 mRNA sequence and found 40 putative MBEs in the 3′-untranslated region (3′-UTR; Extended Data Fig. 9c). To test whether MSI2 binds to the BCAT1 transcripts, we expressed a FLAG-tagged MSI2 protein in K562 cells and performed RNA immunoprecipitation (RIP). FLAG-MSI2 co-precipitated the BCAT1 transcripts with a >1,500-fold enrichment relative to the vector control (Fig. 4a). In contrast, when RIP was performed with a mutant MSI2 protein in which three phenylalanine residues essential for RNA binding were replaced with leucine16, the amount of the BCAT1 mRNA recovered was markedly diminished (Fig. 4a, RBD), indicating that the co-precipitation of BCAT1 transcript requires the RNA binding activity of MSI2. The transcripts for beta-2-microglobulin (B2M) or c-Myc oncogene (MYC) contain only one copy of a putative MBE in their 3′-UTRs (data not shown), and MSI2 RIP did not enrich B2M or MYC mRNAs as efficiently as BCAT1 (Fig. 4a). Furthermore, RIP with an anti-MSI2 antibody showed that endogenous MSI2 proteins bound to BCAT1 transcripts, while B2M or MYC mRNAs exhibited minimal enrichment relative to that of an IgG control (Fig. 4b), indicating that MSI2 is specifically associated with the BCAT1 transcripts. Because MSI2 knockdown reduced the levels of BCAT1 protein and phospho-S6K (Extended Data Fig. 9d), the binding of MSI2 to BCAT1 mRNA positively regulates BCAT1 translation and mTORC1 activation. Importantly, BCAT1 over-expression (Fig. 4c) and BCAA supplementation (Fig. 4d) effectively suppressed the attenuation of the colony-forming ability caused by MSI2 knockdown, with a concomitant increase in pS6K levels in a rapamycin-sensitive manner (Fig. 4e). The levels of AKT phosphorylation were unaffected by shMSI2 (Extended Data Fig. 8c). Collectively, our work presented here demonstrates an essential role for the MSI2-BCAT1 axis in myeloid leukemia and provides a proof-of-principle for inhibiting the BCAA metabolic pathway to regulate CML progression (Fig. 4f).
The up-regulation and functional requirements of BCAT1 have been reported in glioblastoma and in colorectal and breast tumors22,23. Interestingly, Musashi proteins also regulate the same spectrum of cancers including myeloid leukemia18–21,24,25, suggesting a highly conserved role for the MSI-BCAT1 pathway in multiple cancer types. Despite the conservation of this pathway, the metabolic role of BCAT1 seems distinct and dependent on the tissue of origin; in the brain, BCAT1 catalyzes BCAA breakdown and glutamate production to enhance tumor growth in glioblastoma23, whereas BCAT1 promotes BCAA production in leukemia. Mayers et al. recently showed that two different types of tumors, specifically pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung carcinoma (NSCLC)26, exhibit different usages of BCAAs. Despite the same initiating events of Kras activation and p53 deletion, NSCLC cells actively utilize BCAAs by enhancing their uptake and oxidative breakdown to BCKAs, whereas PDAC cells display decreased uptake and thus little dependency on BCAAs. Consistently, BCAT1 and BCAT2 are required for tumor formation in NSCLC but not in PDAC. Although BCAT1 is functionally required for tumor growth in a broad range of malignancies, these reports and our studies highlight the context-dependent role of the BCAT1 metabolic pathway in cancer.
Methods
Mice
C57BL6/J mice were from the Jackson Laboratory. Mice were bred and maintained in the facility of the University Research Animal Resources at University of Georgia. All mice were 8–16 weeks old, age- and sex-matched and randomly chosen for experimental use. No statistical methods were used for sample size estimates. All animal experiments were performed according to protocols approved by the University of Georgia Institutional Animal Care and Use Committee.
Cell isolation, analysis and sorting
Cells were suspended for cell sorting in Hanks’ balanced salt solution (HBSS) containing 5%(vol/vol) fetal bovine serum (FBS) and 2 mM EDTA as previously described27. The following antibodies were used to define lineage positive cells: 145-2C11 (CD3ε), GK1.5 (CD4), 53–6.7 (CD8), RB6-8C5 (Ly-6G/Gr1), M1/70 (CD11b/Mac-1), TER119 (Ly-76/TER119), 6B2 (CD45R/B220), and eBio1D3 (CD19). Red blood cells were lysed with RBC Lysis Buffer (eBioscience) before staining for lineage markers. For the Lin− Sca-1+ cKit+ (LSK) bone marrow cell sorting, the antibodies 2B8 (cKit/CD117) and D7 (Sca-1/Ly-6A/E) antibodies were also used. To determine donor-derived chimerism in the transplantation-based assays, peripheral blood from the recipients was obtained by the submandibular bleeding method and prepared for analysis as previously described20. All antibodies were purchased from eBioscience. Apoptosis assays were performed by staining cells with Annexin V and 7-AAD (BioLegend). Cell cycle status was analyzed by staining cells with 2.5 μg/ml PI containing 0.1% BSA and 2 μg/ml RNase after fixation with 70% ethanol. Flow cytometric analysis and cell sorting were carried out on the Moflo XDP, Cyan ADP (Beckman Coulter) or S3 (Bio-Rad), and the data were analyzed with FlowJo software (Tree Star Inc.).
Viral constructs and production
Retroviral BCR-ABL1 and NUP98-HOXA9 vectors and lentiviral FG12-UbiC-GFP vector were obtained from Addgene. Mouse Bcat1 cDNA (IMAGE clone ID 30063465) was cloned into MSCV-IRES-GFP and Human BCAT1 cDNA (NITE clone ID AK056255) was cloned into FG12-Ubc-hCD2. The short hairpin RNA constructs against Bcat1 (shBcat1) were designed and cloned in MSCV-LTRmiR30-PIG (LMP) vector from Open Biosystems or TtRMPVIR from Addgene according to their instructions. The target sequences are 5′-CCCAGTCTCTGATATTCTGTAC-3′ for shBcat1-a, 5′- TCCGCGCCGTTTGCTGGAGAAA-3′ for shBcat1-b and 5′-CTGTGCCAGAGTCCTTCGATAG-3′ for luciferase as a negative control (shCtrl). Lentiviral short hairpin RNA (shRNA) constructs were cloned in FG12 essentially as described previously28. The target sequences are 5′-CGCAGAGTGTACCGGAGA-3′ for shBCAT1-c, 5′-TGCCCAATGTGAAGCAGT-3′ for shBcat1-d and 5′-TGCGCTGCTGGTGCCAAC-3′ for luciferase as a negative control. Virus was produced in 293FT cells transfected using polyethylenimine with viral constructs along with VSV-G and gag-pol. For lentivirus production Rev was also co-transfected. Viral supernatants were collected for two days followed by ultracentrifugal concentration at 50,000× g for 2h.
Cell culture and colony formation assays
The human BC-CML cell line K562, the human acute leukemia cell lines MV4-11 and U937 were maintained in Roswell Park Memorial Institute 1640 medium (RPMI-1640) with 10% FBS, 100 IU/ml penicillin and 100 μg/ml streptomycin. The human acute promyelocytic leukemia cell line HL60 was maintained in RPMI supplemented with 20% FBS. All human cell lines were obtained from ATCC, and cell line authentication testing was performed by ATCC-standardized STR analysis to verify their identity in July 2016. For the colony forming assays, the cells were transduced with lentiviral shRNA and plated in triplicate in 1.2% methylcellulose medium (R&D systems) supplemented with 100IU/ml penicillin and 100μg/ml streptomycin, 10% FBS. Where indicated, either BCAAs (L-Leucine, L-Valine, L-Isoleucine, 4 mM each, Sigma-Aldrich), L-alanyl-L-glutamine (4 mM, GlutaMax™, Life Technologies), rapamycin (50 nM, Tocris) or gabapentin (Gbp; Tokyo Chemical Industry Co.) was added to the medium. Gbp was freshly dissolved in PBS before use. Colonies were scored on days 9 to 14. For liquid culture of murine cells, freshly isolated adult LSK cells or Lin− BC-CML cells were plated into a 96-well U bottom plate in X-Vivo15 (with Gentamicin and Phenol Red; Lonza) supplemented with 50 μM 2-mercaptoethanol, 10% FBS, 100 ng/ml stem cell factor (SCF, eBioscience) and 20 ng/ml thrombopoietin (TPO, Peprotech). For the BC-CML and LSK colony formation assays, BCR-ABL+ NUP98-HOXA9+ or infected construct-positive cells were sorted and plated in triplicate in Iscove’s modified medium (IMDM)-based methylcellulose medium (Methocult M3434, StemCell Technologies). Colonies were scored on days 7 to 10.
Generation and analysis of leukemic mice
Mice bearing CP- and BC-CML were generated essentially as previously described3,4,29–31. In brief, CP-CML was modeled by transducing the oncogene BCR-ABL1 into hematopoietic stem/progenitor cells (HSPCs) defined by the LSK surface marker phenotype from normal bone marrow, which were transplanted into conditioned recipient mice. BC-CML was modeled by transplanting LSK cells infected with two oncogenes, BCR-ABL1 and NUP98-HOXA9, which are associated with myeloid BC-CML in humans. LSK cells were sorted from healthy C57BL6/J bone marrow and cultured in X-Vivo15 media supplemented with 50 μM 2-mercaptoethanol, 10% FBS, 100 ng/ml SCF and 20 ng/ml TPO. After incubation overnight, cells were infected with retroviruses carrying the oncogenes. Viruses used were as follows: MSCV-BCR-ABL-IRES-YFP to generate CP-CML, or MSCV-BCR-ABL-IRES-YFP and MSCV-NUP98-HOXA9-IRES-tNGFR to generate BC-CML. Cells were harvested 48 h after infection and transplanted retro-orbitally into groups of C57BL6/J mice. Recipients were lethally irradiated (9.5 Gy) for CP-CML and sublethally (6 Gy) for BC-CML. For Bcat1 overexpression, LSK cells were infected with MSCV-BCR-ABL-IRES-YFP and MSCV-Bcat1-IRES-GFP, and doubly infected cells were FACS-purified and transplanted into recipients that were sublethally irradiated. For Bcat1 knockdown by retroviral shRNA transduction, the Lin− population from BC-CML cells was sorted and infected with either control shCtrl (against luciferase) or shBcat1-a/b (against Bcat1) retrovirus for 48 h. Infected cells were sorted based on GFP expression, and 1,000 or 2,000 cells were transplanted in sublethally irradiated C57BL6/J recipients. For conditional Bcat1 knockdown by a Dox-inducible shRNA system, animals were analyzed for donor chimerism at day 10 post-transplantation, and then Dox treatment was initiated by feeding Dox-containing rodent chow (0.2 mg/g diet; S3888, BioServ). After transplantation, recipient mice were maintained on antibiotic water (sulfamethoxazole/trimethoprim) and evaluated daily for signs of morbidity, weight loss, failure to groom, and splenomegaly. Premorbid animals were sacrificed, and relevant tissues were harvested and analyzed by flow cytometry and histopathology. For secondary BC-CML transplantations, cells recovered from terminally ill primary recipients were sorted for Lin− donor cells and transplanted into secondary recipients. Where indicated, sorted live BC-CML cells from the spleen were cytospun and stained with Wright’s stain solution (Harleco) for cytopathologic evaluation by a board-certified veterinary pathologist (T. N.).
Primary human leukemia samples
Patient blood samples were obtained at the Institute of Medical Science, the University of Tokyo (IMSUT) Hospital with written informed consent according to the procedures approved by the Institutional Review Board. Mononuclear cells from the subjects were viably frozen and stored in liquid nitrogen. For in vitro colony formation with BCAT1 knockdown, primary hCD34+ cells sorted from patient bone marrow samples were cultured in IMDM supplemented with 10% FBS, 100 IU/ml penicillin and 100 μg/ml streptomycin, 55 μM 2-mercaptoethanol, SCF, IL-3, IL-6, FLT3L and TPO. After 24 h of culture, the cells were transduced with lentiviral shRNA (cloned in FG12-UbiC-GFP), and the GFP-positive infected cells were sorted at 48 h, and 5,000 - 50,000 cells were plated in complete methylcellulose medium (Methocult H4435, StemCell Technologies). For the colony forming assays with Gbp, sorted hCD34+ cells from the primary patient specimens were cultured in complete methylcellulose medium with the indicated concentrations of Gbp. Colonies were scored on days 9 to 14.
Bioinformatic analysis of human gene expression
For the focused gene expression analysis of BCAT1, BCAT2 and MSI2 in human CML progression, the GEO dataset GSE4170 was retrieved and analyzed using Python v2.7 and the SciPy statistical toolkit (http://www.scipy.org/). Pearson correlation coefficients were used to find patterns of coexpression. For coexpression analysis of BCAT1 and MSI2 across multiple cancer types, the GEO datasets GSE14671 (CML), GSE10327 (medulloblastoma), GSE20916 (colorectal), GSE14548 (breast) and TCGA datasets LAML (AML) and LUAD (lung adenocarcinoma) were collected and analyzed in a similar fashion.
Realtime and standard RT-PCR analysis
Total cellular RNAs were isolated using RNAqueous-Micro kit (Ambion) and cDNAs were prepared from equal amounts of RNAs using Superscript III reverse transcriptase (Life Technologies). For standard PCRs, the reactions were performed with DreamTaq PCR Master Mix (Life Technologies), cDNA and 0.5 μM of each primer. PCR conditions were as follows: 1 min at 94°C, followed by 35 cycles at 94°C for 30 s, 58°C for 30 s, and 72°C for 30 s. PCR primer sequences are as follows. B2m-F1, 5′ –GTAGCCTCTGCCATAGGTTGC-3′; B2m-R2, 5′ –CCATACTGGCATGCTTAACTCTG-3′; Bcat1-F1, 5′ –TGTGGCTGTACGGCAAGGACAAC-3′; Bcat1-R2, 5′ -GTAGCTCGATTGTCCAGTCACT-3′. Quantitative real-time PCRs were performed using EvaGreen® qPCR Master Mix (Bio-Rad) on an iQ5 (Bio-Rad), or using TaqMan Gene Expression Assays on an Applied Biosystems® 7500 Real-Time PCR Systems (Life Technologies). Results were normalized to the level of β-2-microglobulin. PCR primer sequences are as follows. mB2m-F, 5′ –ACCGGCCTGTATGCTATCCAGAA-3′ ; mB2m-R, 5′ –AATGTGAGGCGGGTGGAACTGT-3′; hB2M-F, 5′ –ATGAGTATGCCTGCCGTGTGA-3′; hB2M-R, 5′ –GGCATCTTCAAACCTCCATG-3′; hBCAT1-F, 5′ –TGGAGAATGGTCCTAAGCTG-3′; hBCAT1-R, 5′ –GCACAATTGTCCAGTCGCTC-3′; hMYC-F, 5′ –GAGCAAGGACGCGACTCTCC-3′; hMYC-R, 5′ –GCACCGAGTCGTAGTCGAGG-3′. Following genes were analyzed with TaqMan Gene Expression Assays: Bcat1 (Mm00500289_m1), Bcat2 (Mm00802192_m1), Gpt1 (Mm00805379_g1), Gpt2 (Mm00558028_m1), Got1 (Mm00494698_m1), Got2 (Mm00494703_m1).
Amino acid and keto acid quantification
Leukemia cells or peripheral blood samples drawn from mice bearing myeloid leukemia were used for amino acid and keto acid analysis by high-performance liquid chromatography (HPLC)-fluorescence detection, as described32–34. In brief, two hundred thousand live leukemia cells per sample were sorted and washed twice with ice-cold PBS to remove media components prior to amino acid extraction. The blood plasma was prepared by centrifugation of the peripheral blood samples at 2,000 × g at 4°C for 10 min. Plasma fractions were then treated with 45% methanol/45% acetonitrile containing 6-aminocaproic acid (internal standard for amino acid analysis) or α-ketovalerate (internal standard for keto acid analysis) on ice for 10 min. Cell samples were treated with 80% methanol instead of 45% methanol/acetonitrile mixture. After removing the insoluble particles by centrifugation, the supernatants were collected and dried using a SpeedVac at 30–45°C. For amino acid quantification, the dried samples were treated with the amine-reactive 4-fluoro-7-nitro-2,1,3-benzoxadizole (NBD-F) to derivatize the amino acids. HPLC separation of NBD-amino acids was carried out on an Inertsil ODS-4 column (3.0 × 250 mm, 5 μm, GL Sciences, Tokyo, Japan) at a flow rate of 0.6 ml min−1. We used two types of mobile phase conditions for the separation of 16 amino acids. The mobile phases included (A) 25 mM citrate buffer containing 25 mM sodium perchlorate (pH 6.2) and (B) water/acetonitrile (50/50, v/v). The gradient conditions were as follows: t=0 min, 10% B; t=20 min, 50% B; and t=30 min, 100% B. For NBD-Asn, Ser, Thr, Gln and Phe analysis, 25 mM citrate buffer containing 25 mM sodium perchlorate (pH 4.4) was used as the mobile phase A. NBD-amino acids were detected with excitation and emission wavelengths of 470 and 530 nm, respectively. For keto acid quantification, dried samples were treated with o-phenylenediamine (OPD) to derivatize alpha-keto acids, followed by liquid-liquid extraction with ethyl acetate. HPLC separation of OPD-keto acids was carried out on an Inertsil ODS-4 column (3.0 × 250 mm, 5 μm) at a flow rate of 0.6 ml min−1. Mobile phase was water/methanol (55/45, v/v). The fluorescence detection was carried out at the emission wavelength of 410 nm with excitation of 350 nm.
Measurement of leucine uptake in primary mouse leukemia cells
Primary mouse leukemia cells from the spleens of the mice bearing myeloid leukemia were used for the analysis of leucine uptake essentially as described previously35,36. In brief, live leukemia cells were sorted and washed with HBSS to remove media components. The cells were incubated at 37°C for 1–3 min with pre-warmed HBSS containing 10 μM [(U)-14C]-L-leucine (Moravek Inc., specific activity, 328 mCi/mmol). The cells were subseuently washed twice with cold HBSS and lysed using 100 mM NaOH. The solubilized cell lysates were mixed with the EcoLume liquid scintillation cocktail (MP Biomedicals), and radioactivity was measured using an LS6500 liquid scintillation counter (Beckman Coulter). Leucine uptake was quantified using a calibration curve of [14C]-L-leucine reference standard samples.
NMR-based metabolic analysis
Cells were cultured and labeled in media supplemented with either 170 μM [(U)-13C]-L-valine, 30 or 170 μM [(U)-13C]-ketoisovalerate (KIV) sodium salt (for 13C tracer experiments; Cambridge Isotope Laboratories) or 2 mM [amine-15N]-L-glutamine (for 15N tracer experiments; Cambridge Isotope Laboratories). The concentrations are based on the standard RPMI-1640 media formulation. At the time of collection, the cells were washed twice with ice-cold PBS and extracted with 80% methanol on ice for 10 min. After removing the insoluble particles by centrifugation, the supernatants were collected and dried using a SpeedVac at 30°C. The cell extracts were dissolved in a total volume of 90 μL 99.96% D2O containing 0.1mM DSS-d6 and transferred to 3-mm NMR tubes (Shigemi Inc.). Calibration samples (150–250 mM) were prepared from 98% 15N-enriched glutamine, glutamic acid, valine, leucine, isoleucine and alanine (Isotec Inc.) and 13C-enriched KIV and 13C,15N-enriched valine (Cambridge Isotope Laboratories) in D2O containing 0.1 mM DSS. All signals were identified either with authentic samples or by reference to literature values. Two-dimensional proton correlated spectra (COSY and TOCSY) were also collected in some cases to confirm assignments. The data were collected at 25°C on Agilent DD2 spectrometers at 600 or 900 MHz equipped with cryogenically cooled probes. The 1H data were collected with a 20-sec relaxation delay for accurate integration. The 15N data were acquired with a two-dimensional heteronuclear multiple bond correlation experiment (gNhmbc) derived from the Agilent pulse program library with the transfer delay set for a 15N-1H coupling value of 4 Hz. Typically, data sets were 2000 × 64 complex points with the 15N dimension set between 30 and 46 ppm, and 64 scans per point. The 13C data were acquired with a two-dimensional heteronuclear single bond correlation experiment (HSQCAD) from the Agilent pulse program library, and the datasets were 1202 × 64 complex points with the 13C dimension set between 10 and 80 ppm with 16 scans per point. One-dimentional spectra were also collected using the same heteronuclear correlation experiments for 15N and 13C. The data were processed using MestReNova software (Mestrelab Research S.L.). One-dimensional proton data were processed with 0.3 Hz line broadening and polynomial baseline correction. The gNhmbc and HSQC data were processed with linear prediction and zero-filling in the 15N and 13C dimensions. Integration was achieved by summing over peak areas with the contribution of noise subtracted in the 15N spectra. To calculate the concentrations in the 15N tracer experiments, the 1H and gNhmbc spectra of the calibration samples were integrated, and a scaling factor was derived from the ratio of the known concentration of each 98% enriched 15N-amino acid and the integral values from the gNhmbc data. These factors are a function of the 3-bond coupling between the 15N-amine and β-protons as well as the number of those protons. Therefore, the concentrations of each amino acid in cell extracts can be estimated from their integral values by applying the respective scaling factor. For quantification of 13C-labeled compounds, the methyl groups in the 1H and HSQC spectra of the calibration references were integrated, and a scaling factor was derived essentially as described above and used to calculate concentrations from the HSQC data of each sample.
Antibodies
Anti-FLAG monoclonal antibody M2 (Sigma-Aldrich), anti-MSI2 monoclonal antibody EP1305Y (Abcam) and normal Rabbit IgG PP64B (Millipore) were used for immunoprecipitation. For Western blotting the following antibodies were used: mouse monoclonal BCAT1 (clone ECA39, BD Transduction Laboratories) and Bcat1 OTI3F5 (OriGene), rabbit monoclonal S6K (#9202 and #2708), pS6K (#9234), AKT (#4691), pAKT, T308 (#13038) and pAKT, S473 (#4060) from Cell Signaling, rabbit monoclonal MSI2 EP1305Y, mouse monoclonal HSP90 F-8 (Santa Cruz Biotech) and mouse monoclonal β-tubulin BT7R (Thermo Fisher Scientific).
RNA immunoprecipitation assays
K562 cells were lysed in 50 mM Tris/HCl (pH 7.5) containing 150 mM NaCl, 5 mM EDTA, 1% NP-40, and the Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific). We performed immunoprecipitations with anti-FLAG, anti-MSI2 or rabbit normal IgG and protein G magnetic beads (Life Technologies) for 1 h at 4°C. The immunoprecipitated protein-RNA complexes were washed three times with low- and high-salt wash buffers (300 mM or 550 mM NaCl, respectively), followed by three washes in PBS. Total RNAs were purified from the washed beads using the RNAqueous-Micro kit (Ambion) and subjected to RT-qPCR analysis for quantification. The fold enrichment of the transcript amount in the RIP fraction over the amount present in the input sample before RIP (RIP/input) was calculated for each sample.
Statistical analysis
Statistical analyses were carried out using GraphPad Prism software version6.0f (GraphPad Software Inc.). Data are shown as the mean ± the s.e.m. Two-tailed unpaired Student’s t-tests or Mann-Whitney U tests were used to determine statistical significance. For Kaplan Meier survival analysis, log-rank tests were used for statistical significance (*p<0.05, **p<0.01, ***p<0.001).
Data availability
Source gel images and source data from animal models are included in Supplementary Information files. All NMR spectral data from metabolic analyses are deposited under the Project ID PR000423 in Metabolomics Workbench37. All other relevant data are available from the corresponding author upon request.
Supplementary Material
Extended Data
Acknowledgments
We thank Drs. Warren Pear, David Baltimore and Scott Lowe for plasmids and Drs. Stephen Dalton, Craig Jordan, Bryan Zimdahl, Jun Ninomiya-Tsuji, Kazuhito Sai, Kunihiro Matsumoto, Hiroshi Hanafusa, Tomoaki Mizuno, Yachiyo Kuwatsuka, Yosuke Minami and Matthew Merritt for discussions and comments on the manuscript. We also thank Dr. Julie Nelson at the CTEGD Cytometry Shared Resource Lab for assistance in cell sorting, Drs. Kazuhisa Sekimizu, Christopher West, Msano Mandalasi and Hanke van der Wel for advice on radioisotope use, and Kristen MacKeil, Jamie Nist and Kazutomo Ogata for technical help. This work was supported by grants from the University of Georgia Research Foundation and the Heather Wright Cancer Research Fund (T.I.); by the Japan Society for the Promotion of Science Bilateral Open Partnership Joint Research Projects Program (M.T.); A.S.E. and the CCRC NMR facility were partially supported by the Southeast Center for Integrated Metabolomics, NIH U24DK097209 and the Georgia Research Alliance.
Footnotes
Author Contributions
A.H. designed the studies, performed all experiments, analyzed the data and wrote the manuscript. M.T. designed and performed experiments related to quantitative analysis of amino and keto acids. T.K., M.K. and A.T. provided and performed experiments with human primary samples. T.N. performed histological and cytological analysis. J.G. F.T. and A.S.E. designed and conducted NMR-based metabolic analysis. D.M. and N.K. performed bioinformatics analysis of gene expression datasets. T.I. conceived and supervised the project and wrote the manuscript.
Author Information
Competing financial interests. T.I. and A.H. are named inventors of a provisional patent application number 62/413,028. Correspondence and requests for materials should be addressed to T.I. (ito@bmb.uga.edu).
References
- 1.Vander Heiden MG. Targeting cancer metabolism: a therapeutic window opens. Nat Rev Drug Discov. 2011;10:671–684. doi: 10.1038/nrd3504. [DOI] [PubMed] [Google Scholar]
- 2.DeBerardinis RJ, Thompson CB. Cellular metabolism and disease: what do metabolic outliers teach us? Cell. 2012;148:1132–1144. doi: 10.1016/j.cell.2012.02.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Daley G, Van Etten R, Baltimore D. Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science. 1990;247:824–830. doi: 10.1126/science.2406902. [DOI] [PubMed] [Google Scholar]
- 4.Dash AB, et al. A murine model of CML blast crisis induced by cooperation between BCR/ABL and NUP98/HOXA9. Proc Natl Acad Sci USA. 2002;99:7622–7627. doi: 10.1073/pnas.102583199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Brosnan JT, Brosnan ME. Branched-chain amino acids: enzyme and substrate regulation. J Nutr. 2006;136:207S–11S. doi: 10.1093/jn/136.1.207S. [DOI] [PubMed] [Google Scholar]
- 6.Hutson SM, Sweatt AJ, Lanoue KF. Branched-chain amino acid metabolism: implications for establishing safe intakes. J Nutr. 2005;135:1557S–64S. doi: 10.1093/jn/135.6.1557S. [DOI] [PubMed] [Google Scholar]
- 7.Hutson SM, et al. Role of branched-chain aminotransferase isoenzymes and gabapentin in neurotransmitter metabolism. J Neurochem. 1998;71:863–874. doi: 10.1046/j.1471-4159.1998.71020863.x. [DOI] [PubMed] [Google Scholar]
- 8.Radich JP, et al. Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci USA. 2006;103:2794–2799. doi: 10.1073/pnas.0510423103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wolfson RL, et al. Sestrin2 is a leucine sensor for the mTORC1 pathway. Science. 2016;351:43–48. doi: 10.1126/science.aab2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Han JM, et al. Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell. 2012;149:410–424. doi: 10.1016/j.cell.2012.02.044. [DOI] [PubMed] [Google Scholar]
- 11.Bar-Peled L, Sabatini DM. Regulation of mTORC1 by amino acids. Trends Cell Biol. 2014;24:400–406. doi: 10.1016/j.tcb.2014.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jewell JL, Russell RC, Guan KL. Amino acid signalling upstream of mTOR. Nat Rev Mol Cell Biol. 2013;14:133–139. doi: 10.1038/nrm3522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Okano H, Imai T, Okabe M. Musashi: a translational regulator of cell fate. J Cell Sci. 2002;115:1355–1359. doi: 10.1242/jcs.115.7.1355. [DOI] [PubMed] [Google Scholar]
- 14.Sutherland JM, McLaughlin EA, Hime GR, Siddall NA. The Musashi family of RNA binding proteins: master regulators of multiple stem cell populations. Adv Exp Med Biol. 2013;786:233–245. doi: 10.1007/978-94-007-6621-1_13. [DOI] [PubMed] [Google Scholar]
- 15.Rentas S, et al. Musashi-2 attenuates AHR signalling to expand human haematopoietic stem cells. 2016;532:508–511. doi: 10.1038/nature17665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Imai T, et al. The neural RNA-binding protein Musashi1 translationally regulates mammalian numb gene expression by interacting with its mRNA. Mol Cell Biol. 2001;21:3888–3900. doi: 10.1128/MCB.21.12.3888-3900.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cragle C, MacNicol AM. Musashi protein-directed translational activation of target mRNAs is mediated by the poly(A) polymerase, germ line development defective-2. J Biol Chem. 2014;289:14239–14251. doi: 10.1074/jbc.M114.548271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wang XY, et al. Musashi1 regulates breast tumor cell proliferation and is a prognostic indicator of poor survival. Mol Cancer. 2010;9:221. doi: 10.1186/1476-4598-9-221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Muto J, et al. RNA-Binding Protein Musashi1 Modulates Glioma Cell Growth through the Post-Transcriptional Regulation of Notch and PI3 Kinase/Akt Signaling Pathways. PLoS ONE. 2012;7:e33431. doi: 10.1371/journal.pone.0033431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ito T, et al. Regulation of myeloid leukaemia by the cell-fate determinant Musashi. Nature. 2010;466:765–768. doi: 10.1038/nature09171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kharas MG, et al. Musashi-2 regulates normal hematopoiesis and promotes aggressive myeloid leukemia. Nat Med. 2010;16:903–908. doi: 10.1038/nm.2187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yoshikawa R, et al. ECA39 is a novel distant metastasis-related biomarker in colorectal cancer. World J Gastroenterol. 2006;12:5884–5889. doi: 10.3748/wjg.v12.i36.5884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tönjes M, et al. BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild-type IDH1. Nat Med. 2013;19:901–908. doi: 10.1038/nm.3217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lagadec C, et al. The RNA-binding protein Musashi-1 regulates proteasome subunit expression in breast cancer- and glioma-initiating cells. Stem Cells. 2014;32:135–144. doi: 10.1002/stem.1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li N, et al. The Msi Family of RNA-Binding Proteins Function Redundantly as Intestinal Oncoproteins. Cell Rep. 2015;13:2440–2455. doi: 10.1016/j.celrep.2015.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mayers JR, et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science. 2016;353:1161–1165. doi: 10.1126/science.aaf5171. [DOI] [PMC free article] [PubMed] [Google Scholar]
References - Methods
- 27.Domen J, Cheshier SH, Weissman IL. The role of apoptosis in the regulation of hematopoietic stem cells: Overexpression of Bcl-2 increases both their number and repopulation potential. J Exp Med. 2000;191:253–264. doi: 10.1084/jem.191.2.253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Qin XF, An DS, Chen IS, Baltimore D. Inhibiting HIV-1 infection in human T cells by lentiviral-mediated delivery of small interfering RNA against CCR5. Proc Natl Acad Sci USA. 2003;100:183–188. doi: 10.1073/pnas.232688199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mayotte N, Roy DC, Yao J, Kroon E, Sauvageau G. Oncogenic interaction between BCR-ABL and NUP98-HOXA9 demonstrated by the use of an in vitro purging culture system. Blood. 2002;100:4177–4184. doi: 10.1182/blood-2002-04-1244. [DOI] [PubMed] [Google Scholar]
- 30.Neering SJ, et al. Leukemia stem cells in a genetically defined murine model of blast-crisis CML. Blood. 2007;110:2578–2585. doi: 10.1182/blood-2007-02-073031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ashton JM, et al. Gene Sets Identified with Oncogene Cooperativity Analysis Regulate In Vivo Growth and Survival of Leukemia Stem Cells. Cell Stem Cell. 2012;11:359–372. doi: 10.1016/j.stem.2012.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Song Y, Funatsu T, Tsunoda M. Amino acids analysis using a monolithic silica column after derivatization with 4-fluoro-7-nitro-2,1,3-benzoxadiazole (NBD-F) J Chromatogr B Analyt Technol Biomed Life Sci. 2011;879:335–340. doi: 10.1016/j.jchromb.2010.12.018. [DOI] [PubMed] [Google Scholar]
- 33.Song Y, Funatsu T, Tsunoda M. Rapid determination of amino acids in biological samples using a monolithic silica column. Amino Acids. 2012;42:1897–1902. doi: 10.1007/s00726-011-0914-2. [DOI] [PubMed] [Google Scholar]
- 34.Pailla K, Blonde-Cynober F, Aussel C, De Bandt JP, Cynober L. Branched-chain keto-acids and pyruvate in blood: measurement by HPLC with fluorimetric detection and changes in older subjects. Clin Chem. 2000;46:848–853. [PubMed] [Google Scholar]
- 35.Segawa H, et al. Identification and functional characterization of a Na+-independent neutral amino acid transporter with broad substrate selectivity. J Biol Chem. 1999;274:19745–19751. doi: 10.1074/jbc.274.28.19745. [DOI] [PubMed] [Google Scholar]
- 36.Morimoto E, et al. Establishment and Characterization of Mammalian Cell Lines Stably Expressing Human L-Type Amino Acid Transporters. J Pharmacol Sci. 2008;108:505–516. doi: 10.1254/jphs.08232fp. [DOI] [PubMed] [Google Scholar]
- 37.Sud M, et al. Metabolomics Workbench: An international repository for metabolomics data and metadata, metabolite standards, protocols, tutorials and training, and analysis tools. Nucleic Acids Res. 2016;44:D463–70. doi: 10.1093/nar/gkv1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Source gel images and source data from animal models are included in Supplementary Information files. All NMR spectral data from metabolic analyses are deposited under the Project ID PR000423 in Metabolomics Workbench37. All other relevant data are available from the corresponding author upon request.