Graphical abstract graphic file with name fx1.jpg [49]Open in a new tab Highlights * • Asparagine starvation reduces iron import by downregulating the TFRC * • Iron-dependent histone demethylation is suppressed by asparagine starvation * • IREB2 is responsible for the mRNA expression of TFRC during asparagine starvation * • Metabolic adaptive genes are activated by H3K4 tri-methylation during starvation __________________________________________________________________ Biological sciences; Physiology; Molecular biology; Molecular mechanism of gene regulation; Epigenetics Introduction Mammalian cells respond to amino acid starvation through transcriptional and translational regulation to dictate cell fate.[50]^1^,[51]^2 Recent work suggests that amino acid restriction can also modulate gene expression through altering epigenetic modifications on DNA and histone, thereby impacting cell fate decision.[52]^3^,[53]^4 For example, short-term methionine restriction poises inducible pluripotent stem cells for differentiation by reducing intracellular levels of S-adenosylmethionine (SAM), which is derived from methionine and is an indispensable substrate for DNA and histone methyltransferase.[54]^5 Similarly, studies in multiple mammalian cell systems show that α-ketoglutarate (α-KG) is a key substrate for DNA and histone demethylases.[55]^6^,[56]^7^,[57]^8^,[58]^9^,[59]^10^,[60]^11 Since intracellular α-KG can be maintained through the metabolism of several amino acids, including glutamine, serine, and branch-chained amino acids, manipulating their availability can profoundly affect cell fate decision through modulating histone and DNA demethylation. Unlike the other proteinogenic amino acids, asparagine is the only one that has not been found to be able to be catabolized in mammalian cells,[61]^12 and thus whether its availability can contribute to histone methylation or demethylation has not been studied. Using human leukemic cells that are auxotrophic for asparagine as a model, we reported that asparagine starvation led to an increase in several histone methylation markers. This increase was neither due to the change of expression of histone-modifying enzymes nor due to the alteration of intracellular SAM or α-KG. Instead, asparagine starvation reduces the intracellular pool of labile iron (Fe^2+), an indispensable co-factor for the Jumonji family of histone demethylases (JHDMs), and therefore suppresses histone demethylation. We demonstrate that asparagine starvation suppresses the expression of iron-responsive element-binding protein 2 (IREB2), an iron-sensing protein,[62]^13 to reduce the mRNA expression of the transferrin receptor (TFRC), a major carrier for iron uptake.[63]^14 Furthermore, iron supplementation under conditions of asparagine starvation restored histone demethylation and altered gene expression to accelerate cell death. Taken together, our results identified that regulation of iron-dependent histone demethylation is part of the metabolic adaptation during asparagine starvation. Results Asparagine starvation leads to an increase of histone methylation To determine the impact of asparagine availability on histone methylation status, we withdraw asparagine from the culture medium in four acute lymphoblastic leukemia (ALL) cell lines. We found that depletion of exogenous asparagine for 24–48 h caused a global increase of tri-methylation of histone H3 subunit at multiple lysine residues, including K27, K4, and K9 ([64]Figures 1A and [65]S1A), in RS4; 11 and DND-41 cells that do not express asparagine synthetase (ASNS) ([66]Figure 1B). There was no change of these markers in SEMK2 and Jurkat cells that express high levels of ASNS ([67]Figures 1A and 1B). We also did not observe any change of di-methylation at H3K4 residue in all these four cell lines ([68]Figure 1A). Overexpression of ASNS in RS4; 11 cells prevented the increase of H3K27me3, H3K4me3, and H3K9me2/3 following asparagine depletion ([69]Figure 1C), while ASNS deletion in Jurkat cells induced these 3 markers when exogenous asparagine was removed ([70]Figure 1D). These results suggest that the inability to maintain asparagine availability is the trigger of the global increase of histone H3 methylation. Figure 1. [71]Figure 1 [72]Open in a new tab Asparagine starvation suppresses histone demethylation (A) RS4; 11, DND-41, SEMK2, and Jurkat cells were subjected to asparagine depletion for 24 h. Cellular expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. (B) The expression of ASNS protein was determined by Western blotting in RS4; 11, DND-41, SEMK2, and Jurkat cells. (C) ASNS was overexpressed in RS4; 11 cells through lentivirus-mediated transduction. Control or ASNS-transduced cells were subjected to asparagine depletion for 24 h. The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. (D) ASNS gene was deleted in Jurkat by CRISPR. Control or ASNS-deleted Jurkat cells were subjected to asparagine depletion for 24 h. The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. (E) RS4; 11 cells were subjected to asparagine or glutamine depletion for 24 h. Intracellular levels of α-ketoglutarate (α-KG) and S-adenosylmethionine (SAM) were determined by quantitative LC-MS. (F) RS4; 11 cells were subjected to asparagine or glutamine depletion for 24 h with or without 2 mM Dimethyl-α-ketoglutarate (Dm-α-KG). The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. (G) RS4; 11, DND-41, and Jurkat cells were subjected to the treatment for 24 h and then stained with BioTracker Far-Red for Fe^2+ detection by FACS. DFO was added at 100 μM in the presence of asparagine as a control. (H) Quantification of the mean fluorescence intensity (MFI) of the BioTracker Far-Red staining in the panel (G). (I) RS4; 11 and DND-41 cells were treated with 100 μM DFO in the presence of asparagine for 24 h. The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. (J) RS4; 11 and DND-41 cells were subjected to asparagine depletion for 24 h. Hinokitiol (0.75 μM) with or without Fe(NO[3])[3] (10 μM) was added and the MFI of BioTracker Far-Red was determined by FACS. (K) RS4; 11 and DND-41 cells were treated as panel (J). The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. Results in panels E, H, and J were shown as mean ± SD (standard derivation). p values were determined by using Student’s two-tailed unpaired t-test. See also [73]Figure S1. Since amino acid metabolism can affect intracellular levels of SAM and α-KG, we performed quantitative liquid chromatography mass spectrometry (LC-MS) to measure α-KG and SAM. We found that asparagine starvation reduced α-KG to ∼40%, while glutamine starvation nearly depleted intracellular α-KG ([74]Figure 1E). In addition, asparagine starvation neither increased intracellular SAM or 2-hydroxylglutarate levels nor altered the ratio of α-KG/succinate or SAM/SAH (S-adenosyl-homocysteine) ([75]Figures 1E, [76]S1B and S1C). To determine whether the reduction of α-KG by asparagine starvation causes increased histone H3 methylation, we supplemented cells with a cell-permeable α-KG (Dm-α-KG). We found that Dm-α-KG supplementation reduced H3K27me3, H3K4me3, and H3K9me2/3 under glutamine starvation, but not under asparagine starvation ([77]Figure 1F), suggesting the reduction of α-KG by asparagine starvation is not the cause of increased histone H3 methylation. In addition, we found no correlative changes in the expression of histone methyltransferases or demethylases that could consistently explain the change of these 3 markers during asparagine starvation ([78]Figure S1D). Asparagine starvation reduces the intracellular pool of labile iron (Fe^2+) and thereby suppresses histone demethylation To determine whether histone methyltransferases contributed to the increase of H3 lysine methylation during asparagine starvation, we treated RS4; 11 cells with an inhibitor of EZH2 (GSK126),[79]^15 a known H3K27 methyltransferase. We found that GSK126 was not as effective in reducing the H3K27me3 in the asparagine-depleted conditions, suggesting compromised demethylase activities during asparagine starvation ([80]Figure S1E). Since the demethylation of H3K27me3, H3K4me3, and H3K9me2/3 requires JHDMs that also use Fe^2+ as a co-factor, we measured intracellular Fe^2+ levels using BioTracker Far-red, a dye specific for ferrous.[81]^16 We used deferoxamine (DFO), an iron chelator, to treat the cells in the complete medium as a negative control. We found that asparagine starvation for 24 h reduced Fe^2+ content to less than 50% in RS4; 11 and DND-41 cells, but not in Jurkat cells ([82]Figures 1G and 1H). Consistent with the literature,[83]^17 DFO treatment induced H3K27me3, H3K4me3, and H3K9me2/3 in RS4; 11 and DND-41 cells, with no impact on H3K4me2 ([84]Figure 1I). Since demethylation of H3K4me2 uses LSD1/2, two lysine-specific demethylases that do not use iron as a co-factor,[85]^18 our results are consistent with the hypothesis that the global increase of histone H3 methylation under asparagine starvation is due to a defect of iron-dependent histone demethylation. To determine whether the reduction of intracellular Fe^2+ is necessary for the increase of histone H3 methylation, we treated RS4; 11 and DND-41 cells with Fe^3+ in the presence of Hinokitiol, a chemical carrier for iron import.[86]^19 We found that Fe^3+ plus Hinokitiol, but not Hinokitiol alone, restored intracellular Fe^2+ content and histone H3 demethylation ([87]Figures 1J and 1K). Furthermore, the reduction of H3K27me3 relies on demethylase activities as GSKJ4, an inhibitor of the H3K27 demethylases,[88]^20 prevented this effect following iron supplementation ([89]Figure S1F). Asparagine starvation reduces the cell surface expression of the transferrin receptor (TFRC) To determine the cause of the reduction of intracellular Fe^2+ pool, we analyzed published RNA-seq data from RS4; 11 cells subjected to asparagine starvation[90]^21 and found reduced expression of iron homeostatic genes ([91]Figures 2A and 2B). We confirmed the reduction of the mRNAs of TFRC, STEAP3, SFXN2, and SLC46A1 ([92]Figure S2A), whose major roles are iron uptake and intracellular transport. Consistently, both total cellular expression and cell surface expression of TFRC, also known as CD71, were reduced by asparagine starvation in RS4; 11 and DND-41 cells, but not in SEMK2 and Jurkat cells ([93]Figures 2C–2E). Furthermore, ASNS overexpression restored TFRC cell surface expression in RS4; 11 cells when asparagine was withdrawn, while ASNS deletion in Jurkat cells reduced TFRC cell surface expression under the same condition ([94]Figures S2B and S2C). Since TFRC is a major carrier for iron uptake,[95]^14 we hypothesize that the reduction of TFRC expression by asparagine starvation leads to the reduction of intracellular Fe^2+ to suppress iron-dependent histone demethylation. To test this hypothesis, we used doxycycline-inducible short hairpin RNA (shRNA) to suppress TFRC expression ([96]Figures 2F and [97]S2D) and found that doxycycline treatment induced H3K27me3 and H3K4me3 in the presence of exogenous asparagine ([98]Figure 2G). This induction was mitigated by iron supplementation in the presence of Hinokitiol ([99]Figure 2G), which correlated with intracellular Fe^2+ content ([100]Figure S2E). Conversely, TFRC overexpression in RS4; 11 and DND-41 cells restored cell surface expression of TFRC, intracellular Fe^2+ content, and partially rescued histone demethylation under asparagine depletion ([101]Figures S2F, S2G, and [102]2H). These results suggest that TFRC is both necessary and sufficient for iron-dependent histone demethylation. Figure 2. [103]Figure 2 [104]Open in a new tab Asparagine starvation reduces intracellular iron content by downregulating the transferrin receptor (TFRC/CD71) (A) Gene set enrichment analysis identified downregulation of mRNAs of iron metabolism genes in RS4; 11 cells following asparagine depletion for 24 h. (B) Heatmap illustration of the change of expression of iron metabolism genes in panel (A). (C) RS4; 11, DND-41, SEMK2, and Jurkat cells were subjected to asparagine depletion for 24 h. TFRC protein expression was determined by Western blotting. (D) RS4; 11, DND-41, SEMK2, and Jurkat cells were subjected to asparagine depletion for 24 h. Cell surface expression of TFRC/CD71 was determined by FACS analysis. (E) The MFI of cell surface staining of TFRC/CD71 in panel (D) was quantified. Results were shown as mean ± SD (standard derivation). p values were determined by using Student’s two-tailed unpaired t-test. (F) The expression of TFRC in RS4; 11 cells was suppressed by doxycycline-inducible shRNA following 48 h of doxycycline (1 μg/mL) treatment. (G) RS4; 11 cells with control or TFRC shRNA were treated with doxycycline for 48 h with or without Hinokitiol (0.75 μM) plus Fe(NO[3])[3] (10 μM). The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. (H) TFRC cDNA was introduced into RS4; 11 and DND-41 via lentivirus-mediated gene transduction. Control or TFRC-transduced cells were subjected to asparagine depletion for 24 h. The expression of H3K27me3, H3K4me3, H3K9me2/3, and H3K4me2 was determined by Western blotting. See also [105]Figure S2. Downregulation of IREB2 is responsible for the reduction of TFRC expression following asparagine depletion The stability of TFRC mRNA is tightly regulated by iron-sensing proteins. ACO1 and IREB2 are the two known iron-sensing proteins that bind to the iron-responsive elements (IREs) within the 3′ UTR of TFRC mRNA to increase its stability when the intracellular iron content is low.[106]^13 Elevated cellular iron inactivates ACO1 and IREB2 via different molecular machinery to cause their dissociation from TFRC mRNA, leading to its degradation and therefore providing a feedback mechanism to maintain iron homeostasis. We found that, unlike TFRC, the mRNA levels of ACO1 and IREB2 did not change following asparagine depletion in both RS4; 11 and DND-41 cells ([107]Figure 3A). However, the protein expression of IREB2 was consistently reduced by asparagine starvation in RS4; 11 and DND-41 cells, but not in SEMK2 and Jurkat cells ([108]Figure 3B). Similarly, only the IREB2 protein, but not ACO1, was reduced by asparagine starvation in Jurkat cells lacking ASNS gene ([109]Figure 3C). Using the doxycycline-inducible shRNAs, we found that only suppression of IREB2 reduced the expression of TFRC at both mRNA and protein levels ([110]Figures S3, [111]3D, and 3E). Figure 3. [112]Figure 3 [113]Open in a new tab Asparagine starvation downregulates the expression of IREB2 post-transcriptionally (A) The mRNA expression of TFRC, ACO1, and IREB2 was determined by qPCR in RS4; 11 and DND-41 cells following 24-h asparagine depletion. (B) RS4; 11, DND-41, SEMK2, and Jurkat cells were subjected to asparagine depletion for 24 h. The expression of TFRC, ACO1, and IREB2 protein was determined by Western blotting. (C) Control or ASNS-deleted Jurkat cells were subjected to asparagine depletion for 24 h. The expression of TFRC, ACO1, and IREB2 protein was determined by Western blotting. (D) RS4; 11 cells were transduced with two inducible hairpins of ACO1 and then treated with doxycycline (1 μg/mL) for 48 h. The expression of ACO1 and TFRC protein was determined by Western blotting. (E) RS4; 11 cells were transduced with two inducible hairpins of IREB2 and then treated with doxycycline (1 μg/mL) for 48 h. The expression of IREB2 and TFRC protein was determined by Western blotting. See also [114]Figure S3. Asparagine starvation increases H3K4me3 deposition to alter gene expression and dictate cell fate Since histone methylation profoundly affects gene expression, we hypothesize that increased H3 lysine methylation under asparagine starvation modulates gene expression to mitigate stress. Using H3K4me3 ChIP-seq, we found a global increase in its deposition across the genome following asparagine depletion ([115]Figure S4A). The enrichment of H3K4me3 on gene promoters was positively correlated with gene expression, consistent with the known role of H3K4me3 on transcriptional activation ([116]Figure S4B). Furthermore, supplementation of Fe^3+ in the presence of Hinokitiol reduced the H3K4me3 deposition in the promoter regions of a portion of genes whose H3K4me3 deposition increased in the promoter regions under asparagine starvation ([117]Figure S4C). There was a weak correlation between the reduction of H3K4me3 deposition and gene expression alteration ([118]Figure S4C). The genes with increased mRNA expression or increased H3K4me3 deposition upon asparagine starvation were significantly enriched in the cell periphery, plasma membrane synapse, and vesicle trafficking given either ChIP-seq ([119]Figure 4A) or RNA-seq results ([120]Figure 4B), indicating a potential enhancement of exchanging with the extracellular environment. For example, genes in the cytoplasmic vesicle membrane pathway were induced by asparagine starvation ([121]Figure 4C). The functions of these genes are broadly linked to membrane internalization, endocytosis, and endosomal function, which can facilitate the utilization of extracellular protein as a source of amino acid to mitigate the stress.[122]^22 In addition, Fe^3+ supplementation suppressed the induction of some of these genes involved in cell surface receptor recycling or vesicle trafficking, such as SLA and FGD2[123]^23^,[124]^24 ([125]Figure 4D, left panel). Furthermore, the mRNA expression of SLA and FGD2 positively correlated with H3K4me3 deposition in their promoter regions under these three conditions ([126]Figure 4D, right panel). Figure 4. [127]Figure 4 [128]Open in a new tab Asparagine starvation leads to an increase of H3K4me3 deposition to modulate gene expression (A) RS4; 11 cells were starved for asparagine for 24 h. H3K4me3 ChIP-seq analysis was performed (n = 3). The differential expression peaks (FDR<0.05, |log2FC|>0.25) were subjected to gene ontology and pathway functional analysis by using DAVID. (B) RS4; 11 cells were starved for asparagine for 24 h. RNA-seq analysis was performed (n = 3). The differential expression genes (FDR<0.05, |log2FC|>0.25) were subjected to gene ontology and pathway functional analysis by using DAVID. (C) RS4; 11 cells were starved for asparagine for 24 h. RNA-seq analysis was performed (n = 3). A heatmap of the genes in the cytoplasmic vesicle membrane pathway was shown. (D) RS4; 11 cells were subjected to asparagine starvation with or without Fe^3+ supplementation for 24 h. RNA was harvested and the relative expression of SLA and FGD2 were determined by qPCR (left). H3K4me3 ChIP-seq analysis was performed under the same conditions and relative enrichment of H3K4me3 around gene promoters was illustrated by the genome browser (right). The result is a merged peak of 3 replicates. (E) RS4; 11 cells were subjected to asparagine starvation for 24 h in the presence of other treatments as shown in the legend. Cell death index was measured by Annexin V staining. (F) RS4; 11 and DND-41 cells were cultured in asparagine-replete or -deficient media for 24 h. Pitstop (50 μM) or EIPA (25 μM) was added in both conditions. Cell death index was measured by trypan blue staining. Results in panels E and F were shown as mean ± SD (standard derivation). p values were determined by using Student’s two-tailed unpaired t-test. See also [129]Figure S4. Previous work from our lab showed that long-term asparagine starvation triggered apoptosis, while short-term (<24 h) asparagine starvation induced growth arrest with minimal cell death.[130]^25 Thus, we hypothesize that cells may uptake extracellular proteins as a means to regenerate intracellular amino acids to transiently mitigate asparagine starvation. Consistent with this hypothesis, Fe^3+ supplementation enhanced cell death within 24 h following asparagine withdrawal ([131]Figure 4E). The enhanced cell death is not due to ferroptosis,[132]^26 as we did not observe significantly increased reactive oxygen species or lipid peroxide ([133]Figures S4D and S4E). Furthermore, Q-VD, a pan-caspase inhibitor, suppressed the enhanced cell death triggered by Fe^3+ supplementation ([134]Figure 4E). To examine our hypothesis, we treated RS4; 11 and DND-41 cells with inhibitors of endocytosis (Pitstop 2) or macropinocytosis (EIPA). The results showed an enhanced cell death under asparagine starvation ([135]Figure 4F). Discussion We conclude that asparagine starvation in asparagine auxotrophic cells inhibits the expression of TFRC and thus suppresses iron uptake and iron-dependent histone demethylation. Iron supplementation can restore histone demethylation and therefore alter gene expression under asparagine starvation. Our results indicate that inhibition of histone demethylation is part of the metabolic adaptive response to ensure proper gene expression to mitigate the stress, as iron supplementation accelerates cell death under asparagine starvation. We identified that downregulation of IREB2 is a key molecular component to be responsible for asparagine starvation-induced inhibition of TFRC expression. However, the mechanism underlying the post-transcriptional inhibition of IREB2 expression by asparagine starvation remains to be determined. For example, FBXL5 is a known E3 ubiquitin ligase of IREB2,[136]^27^,[137]^28 whether its activity is regulated by asparagine starvation warrants further investigation. Since the connection between amino acid restriction and epigenetic regulation only comes to focus recently,[138]^29^,[139]^30 our study will provide a comprehensive understanding of nutrient availability and histone modifications. Furthermore, the dynamic change of TFRC/CD71 expression in a variety of physiological and pathological conditions may indicate a requirement for iron-dependent histone demethylation to regulate epigenome.[140]^31^,[141]^32 Therefore, our results uncover that iron homeostasis is another layer of control between amino acid restriction, epigenome, and cell fate decision. Limitations of the study This study was restricted to in vitro cell culture system. In addition, the phenotype is specific to cells expressing low levels of ASNS. Thus, it is important to determine whether our findings contribute to the phenotype when ALL cells are treated with L-asparaginase in vivo. Furthermore, it is still unclear whether the reduction of intracellular iron content is specific to asparagine starvation or can be responsive to other nutrient restriction or stress conditions. Finally, JHDMs use ferrous (Fe^2+) as a co-factor and our iron detection method can only detect ferrous. Since TFRC-mediated iron import is through transferrin-bound ferric (Fe^3+), it remains to be determined that whether ferric reduction to ferrous is also regulated by amino acid starvation. These questions imply that mammalian cells may regulate iron homeostasis at multiple layers during amino acid starvation to ensure proper iron-dependent metabolic activities to mitigate nutrient stress for cellular adaptation. STAR★Methods Key resources table REAGENT or RESOURCE SOURCE IDENTIFIER Antibodies __________________________________________________________________ Rabbit polyclonal α-ASNS ProteinTech Cat. # 14681-1-AP; RRID: AB_2060119 Rabbit monoclonal α-TFRC/CD71 Cell Signaling Technology Cat. # 13208S; RRID: AB_2798150 APC α-TFRC/CD71 (Flow Cytometry) BioLegend Cat. # 334108; RRID AB_10915138 Rabbit polyclonal α-ACO1 ProteinTech Cat. # 12406-1-AP; RRID: AB_10642942 Rabbit monoclonal α-IREB2 Cell Signaling Technology Cat. # 37135S; RRID: AB_2799110 Rabbit monoclonal α-Tri-Methyl-Histone H3 (K27) Cell Signaling Technology Cat. # 9733T; RRID: AB_2616029 Rabbit polyclonal α-Tri-Methyl-Histone H3 (K4) Cell Signaling Technology Cat. # 9727S; RRID: AB_561095 Rabbit polyclonal α-Tri-Methyl-Histone H3 (K4) (ChIP) Active Motif Cat. # 39016 Rabbit monoclonal α-Di-Methyl-Histone H3 (K4) Cell Signaling Technology Cat. # 9725S; RRID: AB_10205451 Mouse monoclonal α-Di/Tri-Methyl-Histone H3 (K9) Cell Signaling Technology Cat. # 5327T; RRID: AB_10695295 Rabbit polyclonal α-Histone H3 ProteinTech Cat. # 17168-1-AP; RRID: AB_2716755 Mouse monoclonal α-MLL Santa Cruz BioTech Cat. # sc-377274; RRID: AB_2616342 Mouse monoclonal α-EZH2 Santa Cruz BioTech Cat. # sc-515817 Mouse monoclonal α-EZH1 Santa Cruz BioTech Cat. # sc-166609; RRID: AB_2246791 Mouse monoclonal α-KDM6A Santa Cruz BioTech Cat. # sc-514859 Rabbit polyclonal α-KDM5A ABclonal Technology Cat. # A7238; RRID: AB_2767786 Mouse monoclonal α-β-actin (clone AC-74) Sigma-Aldrich Cat. # A2228; RRID: AB_476697 Mouse monoclonal α-α-tubulin (clone DM-1A) Sigma-Aldrich Cat. # T9026; RRID: AB_477593 __________________________________________________________________ Chemicals, peptides, and recombinant proteins __________________________________________________________________ Dimethyl alpha-ketoglutarate Sigma Cat. # 349631 Iron (Fe^3+) nitrate nonahydrate Sigma Cat. # F8508 Deferoxamine mesylate salt Sigma Cat. # D9533 Hinokitiol Sigma Cat. # 469521 doxycycline hydrochloride Sigma Cat. # D3447 Q-VD-OPH SelleckChem Cat. # S7311 GSK126 SelleckChem Cat. # S7061 GSKJ4 SelleckChem Cat. # S7070 Pitstop 2 MedChemExpress Cat. # HY-115604 EIPA MedChemExpress Cat. # HY-101840 BioTracker Far-Red Labile Fe^2+ dye Millipore Sigma Cat. # SCT037 Fixable viability dye eFluor 450 eBioscience Cat. # 65-0863-14 H2DCFDA ROS dye Thermo Fisher Scientific Cat. # D399 LiperFluo Dojindo Molecular Technologies Cat. # [142]L24810 Erastin SelleckChem Cat. # S7242 Tert-Butyl hydroperoxide (t-BHP) Thermo Fisher Scientific Cat. # AC180340050 __________________________________________________________________ Deposited data __________________________________________________________________ RNA seq of leukemia cells following asparagine depletion Jiang et al.[143]^21 GEO: [144]GSE135420 RNA seq of leukemia cells (RS4;11) following asparagine depletion This paper GEO: [145]GSE206112 H3K4me3 ChIP seq of leukemia cells (RS4;11) following asparagine depletion This paper GEO: [146]GSE206112 __________________________________________________________________ Experimental models: Cell lines __________________________________________________________________ RS4;11 ATCC ATCC® CRL-1873™; RRID: CVCL_0093 RS4;11 (control and ASNS overexpressing cells) Jiang et al.[147]^21 NA SEMK2 Ross Levine, MSKCC RRID: CVCL_S906 Jurkat ATCC ATCC® TIB-152™; RRID: CVCL_0065 Jurkat (control and ASNS knockout cells) Srivastava et al.[148]^25 NA DND-41 Hui Feng, Boston University RRID: CVCL_2022 DND-41 (control and ASNS overexpressing cells) Jiang et al.[149]^21 NA __________________________________________________________________ Oligonucleotides __________________________________________________________________ RT-PCR primers See [150]Table S1 NA sgGFP guide RNA sequence GTGAACCGCATCGAGCTGAA IDT DNA NA sgASNS guide RNA sequence TTGTCATAGAGGGCGTGCAG IDT DNA NA shCtrl hairpin RNA sequence CAGGAATTATAATGCTTATCTA IDT DNA NA shTFRC hairpin RNA sequence AGGTGATCATAGTTGATAAG IDT DNA NA shACO1-1 hairpin RNA sequence CGGCAACGATATAAGTACAT IDT DNA NA shACO1-2 hairpin RNA sequence CCCACAGGATACTCTTGAGA IDT DNA NA shIREB2 -1 hairpin RNA sequence CTCAGACCATTTTGTAAATA IDT DNA NA shIREB2 -2 hairpin RNA sequence CGAGTAGCAGATTGAAATAT IDT DNA NA __________________________________________________________________ Recombinant DNA __________________________________________________________________ Lenti-CRISPRv2-Puro-sgCtrl[151]^33 Addgene (Deposited by Brett Stringer) Addgene #98290; RRID: Addgene_98290 Lenti-CRISPRv2-Puro-sgASNS Addgene backbone #98290 Addgene #98290; RRID: Addgene_98290 LeGO-iG2[152]^34 Addgene (Deposited by Boris Fehse) Addgene #27341; RRID: Addgene_27341 LeGO-ASNS-iG2 (ASNS cDNA purchased from Dharmacon) Addgene backbone #27341 Addgene #27341; RRID: Addgene_27341 Second-generation lentiviral vector expressing EGFP and puromycin resistant gene[153]^35 Layer et al.[154]^35 (Shared by Utpal Dave) NA Second-generation lentiviral vector expressing EGFP and puromycin resistant gene, containing TFRC cDNA Layer et al.[155]^35 (Shared by Utpal Dave) NA LT3GEPIR-shCtrl[156]^36 Addgene (Deposited by Johannes Zuber) Addgene #111177; RRID: Addgene_111177 LT3GEPIR-shTFRC Addgene backbone #111177 Addgene #111177; RRID: Addgene_111177 LT3GEPIR-shACO1 Addgene backbone #111177 Addgene #111177; RRID: Addgene_111177 LT3GEPIR-shIREB2 Addgene backbone #111177 Addgene #111177; RRID: Addgene_111177 pMD2.G VSV-G envelope plasmid Addgene (Deposited by Didier Trono) Addgene #12259; RRID: Addgene_12259 __________________________________________________________________ Software and algorithms __________________________________________________________________ Maven[157]^37^,[158]^38 Melamud et al.; Clasquin et al.[159]^37^,[160]^38 [161]http://maven.princeton.edu/index.php; RRID:SCR_022491 STAR (v2.7.2a)[162]^39 Dobin et al.[163]^39 [164]http://code.google.com/p/rna-star/; RRID:SCR_004463 featureCounts (v1.6.2)[165]^40 Liao et al.[166]^40 [167]http://bioinf.wehi.edu.au/featureCounts/; RRID:SCR_012919 edgeR (v3.36.0)[168]^41 McCarthy et al.; Robinson et al.[169]^41^,[170]^42 [171]http://bioconductor.org/packages/edgeR/; RRID:SCR_012802 DAVID[172]^43^,[173]^44 Dennis et al.; Huang da et al.[174]^43^,[175]^44 [176]https://david.ncifcrf.gov/; RRID:SCR_001881 MsigDB (v6)[177]^45 Subramanian et al.[178]^45 [179]http://software.broadinstitute.org/gsea/msigdb/index.jsp; RRID:SCR_016863 Bowtie2[180]^46 Langmead and Salzberg,[181]^46 [182]http://bowtie-bio.sourceforge.net/bowtie2/index.shtml;RRID:SCR_016 368 Picard NA [183]http://broadinstitute.github.io/picard/; RRID:SCR_006525 MACS2[184]^47 Zhang et al.[185]^47 [186]https://github.com/macs3-project/MACS; RRID:SCR_013291 ENCODE blacklist[187]^48^,[188]^49 Amemiya et al.; Consortium,[189]^48^,[190]^49 [191]https://github.com/Boyle-Lab/Blacklist/ [192]Open in a new tab Resource availability Lead contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Ji Zhang ([193]jzh1@iu.edu). Material availability All reagents will be made available on request after the completion of a Material Transfer Agreement. Experimental model and subject details Cell culture All human leukemic cell lines were cultured at 37°C in 5% CO[2] in a lymphocyte culturing medium (LCM). The base of LCM is a 1:1 ratio mixture of DMEM and IMDM. For practical reason and quality control purpose, we can consistently generate LCM in lab through supplementing high glucose DMEM (11965092, Thermo Fisher Scientific) with: 0.14 mM L-Alanine, 0.17 mM L-Proline, 5.3 × 10^5 mM Biotin, 9.59 × 10^−5 mM Vitamin B12 and 9.8 × 10^−5 mM Sodium Selenite, 10 mM HEPES, and 55 μM β-mercaptoethanol, 100 Units/mL penicillin/streptomycin, 2 mM L-Glutamine, 10% FBS and 0.1 mM L-Asparagine. For asparagine or glutamine starvation, cells were centrifuged and supernatant was removed by aspiration. Cell pellets were resuspended in asparagine- or glutamine-free LCM. Asparagine- or glutamine-free LCM was made from high glucose DMEM lacking glutamine (11960044, Thermo Fisher Scientific) as described above, with the exception of using 10% dialyzed FBS. Cell viability and viable cell numbers were recorded in using the Vi-CellXR cell viability analyzer (Beckman Coulter). When the earlier apoptotic marker was measured, we used Annexin V-FITC (BD Bioscience, 560931). Method details Western blotting For non-histone blots, protein was extracted by using 1× RIPA buffer (diluted from 10x RIPA lysis buffer, Millipore, Cat. # 20-188) with protease inhibitors (Thermo Scientific, Cat. # 1860932) and phosphatase inhibitors (Thermo Scientific, Cat. # 78428). For histone blots, cells were first lysed with Triton Extraction Buffer (PBS containing 0.5% Triton plus protease inhibitors). Nuclei were collected by centrifugation and then lysed with 0.2 N HCl overnight at 4°C. Supernatant was collected by centrifugation and then neutralized with 1/10 volume of 2 M NaOH. Total proteins of equal amount (20 μg for non-histone and 5 μg for histone) were separated on NuPAGE Bis-Tris gels (Invitrogen, Cat. # NP0322BOX) and then transferred to Nitrocellulose membranes (Bio-Rad, Cat. # 1620115). Membranes were blocked in 5% milk and then incubated with corresponding primary antibodies overnight at 4°C. Membranes were washed with 1×Tris Buffered Saline with Tween 20/TBST (diluted from 20x TBST, Santa Cruz Biotechnology, Cat. # 362311) and then incubated with horseradish peroxidase (HRP) conjugated secondary antibody (ECL anti-rabbit IgG, Sigma, Cat. # NA934V; ECL anti-mouse IgG, Sigma, Cat. # NA931V. 1:5000 dilution). Membranes were washed with 1× TBST and subjected to Chemiluminescent Western ECL detection (Thermo Scientific, Cat. # 32106). The blots were stripped with Restore Western Blot Stripping Buffer (Thermo Scientific, Cat. # 21059), washed with 1xTBST, and then re-probed with appropriate primary antibodies for signal detection. Mass spectrum analysis of intracellular metabolite Cells were collected by centrifugation, supernatant was aspirated and the pellets were washed thoroughly once with ice cold 1 × HBSS (14025092, Life Technology). Cellular metabolites were extracted with 80% methanol on ice. Supernatant was collected and dried with SpeedVac (SPD111V, Thermo Fisher Scientific) connected to Refrigerated Vapor Trap (RVT5105, Thermo Fisher Scientific) at room temperature. Dried samples were resuspended in 40:40:20 acetonitrile:MeOH:water and analyzed using a Thermo Q-Exactive mass spectrometer coupled to a Vanquish Horizon UHPLC. Metabolites were separated on a 150 × 2.1 mm Xbridge BEH Amide (2.5 μM) HPLC Column (Waters). Samples were run with a gradient of solvent A (95% H[2]O, 5% ACN, 20 mM NH[4]AC, 20 mM NH[4]OH) and solvent B (20% H[2]O, 80% ACN, 20 mM NH[4]AC, 20 mM NH[4]OH) as follows: 0 min, 100% B; 3 min, 100% B; 3.2 min, 90% B; 6.2 min, 80% B; 10.5 min, 80% B, 10.7 min, 70% B; 13.5 min, 70% B; 13.7 min, 45% B; 16 min, 45% B; 16.5 min 100% B; 22 min, 100% B. Data were collected on a full scan. Flow rate was 0.2 ml/min. Metabolites were identified based on exact M/z and retention time determined using chemical standards. Data were analyzed with Maven,[194]^37^,[195]^38 and normalized to internal standard of ^13C[4],^15N[2]-Asparagine (1 pmole/sample) and then total cell number of each sample. Intracellular ferrous (Fe^2+), ROS, and lipid peroxide measurement Cells were collected by centrifugation and washed once with PBS. BioTracker Far-Red (SCT037, Millipore Sigma) was diluted in serum-free DMEM at final concentration of 5 μM. Resuspend 1 million cells in 100 μL of diluted BioTracker Far-Red staining medium in 96-well plates and incubate the plate reaction in 37°C CO[2] incubator for 1 hour. Cells with staining medium was further diluted with 400 μL serum-free DMEM containing eFluor 450 (65-0863-14, eBioscience) at 1:4000 dilution for dead cell labeling. The signal of BioTracker Far-Red was captured in RL1 channel by the Invitrogen Attune NxT Flow Cytometer. For the ROS measurement, cells were stained in 100 μL serum-free DMEM with 5 μM H[2]DCFDA (D399, Thermo Fisher Scientific) at 37°C for 30 minutes and then diluted with 400 μL serum-free DMEM before detection. The signal of H[2]DCFDA was captured in BL1 channel by the Invitrogen Attune NxT Flow Cytometer. Lipid peroxide was detected by Liperfluo (Dojindo Molecular Technologies, [196]L24810) staining at 1 μM for 30 minutes prior to Flow Cytometry analysis. We used APC anti-human CD71 (334108, BioLegend Inc.) to label cell surface expression of CD71. Molecular cloning and virus production Mouse ASNS cDNA was cloned into the LeGO-iG2 vector backbone for overexpression in RS4;11 cells (Addgene, #27341). Human TFRC/CD71 cDNA was cloned into a second-generation lentiviral vector expressing EGFP and puromycin resistant gene at the same time for dual-selection.[197]^35 Guide RNAs for human ASNS gene were designed using Feng Zhang lab’s CRISPR design resource: [198]http://crispor.tefor.net/ and cloned into pLentiCRISPRv2-Puro vector (Addgene, Cat. # 98290). The design of the doxycycline-inducible shRNAs targeting human TFRC, ACO1 and IREB2 was previously described.[199]^50 The hairpins were cloned into the LT3GEPIR vector (Addgene, Cat# 111177).[200]^36 We used pMD2.G (Addgene, Cat# 12259) and psPAX2 (Addgene, Cat# 12260) as packaging plasmids for lentiviral particle production in 293T cells. mRNA quantification Total RNA was then isolated with TRIzol (15596026, Life Technologies) according to the manufacturer’s instructions. 0.5–2 μg total RNA was processed for cDNA synthesis with random hexamer primers, using EasyScript Plus RTase from the EasyScript Plus cDNA Synthesis Kit (Lamda Biotech, Cat. #G235). The synthesized cDNA was then subjected for qPCR amplification with designed PCR primers for human target genes. RNA sequencing Total RNA was first evaluated for its quantity, and quality, using Agilent Bioanalyzer 2100. For RNA quality, a RIN number of 7 or higher is desired. One hundred nanograms of total RNA was used. cDNA library preparation included mRNA purification/enrichment, RNA fragmentation, cDNA synthesis, ligation of index adaptors, and amplification, following the KAPA mRNA Hyper Prep Kit Technical Data Sheet, KR1352 – v4.17 (Roche Corporate). Each resulting indexed library was quantified and its quality accessed by Qubit and Agilent Bioanalyzer, and multiple libraries pooled in equal molarity. The pooled libraries were then denatured, and neutralized, before loading to NovaSeq 6000 sequencer at 300pM final concentration for 100b paired-end sequencing (Illumina, Inc.). Approximately 30–40M reads per library was generated. A Phred quality score (Q score) was used to measure the quality of sequencing. More than 90% of the sequencing reads reached Q30 (99.9% base call accuracy). Chromatin immunoprecipitation followed by high throughput sequencing (ChIP-seq) 1 × 10^7 cells were resuspended in 25 mL PBS and fixed with 1% formaldehyde (final concentration) on a platform rocker at room temperature for 10 min 1.4 mL of 2.5M glycine was added and incubated for another 5 min on platform rocker to quench the crosslinking reaction. Cells were washed and lysed in 2 mL cell lysis buffer A (20 mM Tris-HCl pH = 8.0, 85 mM KCl, and 0.5% NP-40) and incubated on ice for 10 min. Nuclei was pelleted by centrifugation at 1,350 g for 5 min at 4°C. The nuclei was then resuspended in 750 μl lysis buffer B (50 mM Tris-HCl pH = 8.0, 10 mM EDTA, 1% SDS, plus protease inhibitor cocktail) and sonicated by using the Covaris S2 Focused-ultrasonicator until majority of DNA fragments were between 200 and 500 base pairs in size. The sonicated materials were then centrifuged at 20,000 g for 10 min at 4°C to collect supernatant. 10% of the supernatant (75 μL) was used as input and 300 μL supernatant was used for each immunoprecipitation (IP) reaction. The 300 μL supernatant sample was diluted 5-fold by adding 1.2 ml IP dilution buffer (1.25% Triton X-100, 187.5 mM NaCl, 20 mM Tris-HCl pH = 8.0, plus protease inhibitor cocktail), and incubated overnight at 4°C by rotating with the 30 μL protein G Dynabeads (Life Technologies) pre-incubated with H3K4me3 antibody (Active Motif, 39016). The beads were washed consecutively with 1 mL Low-salt wash buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl pH 8.0, and 150 mM NaCl, twice), High-salt wash buffer (0.1% SDS, 1% Triton X-100, 2 mM EDTA, 20 mM Tris-HCl pH 8.0, 500 mM NaCl, once), LiCl wash buffer (0.25 M LiCl, 1% NP-40, 1% Sodium Deoxycholate, 1 mM EDTA, 10 mM Tris-HCl pH 8.0, once) and TE wash buffer (50 mM NaCl, 10 mM Tris pH 8.0, 1 mM EDTA, once) and incubated with 125 μL Elution Buffer (1% SDS/0.1 M sodium bicarbonate) for 15min on thermomixer at 1000 RPM and 65°C. Supernatant was collected by magnet separation from the beads. 5 μL 5M NaCl was added to each elute and incubated at 65°C overnight. 30 μL of input samples were incubated with 95 μL Elution Buffer and 5 μl 5M NaCl was used for reverse crosslinking. 2 μl of RNase A (0.5 mg/mL) was added to each IP and input sample for 30 min incubation at 37°C. 2 μL of Proteinase K (20 mg/mL) was then added to each sample and incubated for 2 h at 55°C. PCR Purification Kit (Qiagen) was used to recover DNA from each sample in 40 μL EB buffer. Traces of eluted DNA were used to library preparation using Illumina TruSeq Nano DNA LT Library Prep Kit (Cat# FC-121-4001), including end-repair, dA-tailing, indexed adaptor ligation and amplification. Each resulting indexed library were quantified and its quality accessed by Qubit and Agilent Bioanalyzer, and multiple libraries were pooled in equal molarity. The pooled libraries were then denatured, and neutralized, before loading onto NovaSeq 6000 sequencer at 300pM final concentration for 100b paired-end sequencing (Illumina, Inc.). Approximately 15-20M reads per library was generated. A Phred quality score (Q score) was used to measure the quality of sequencing. More than 90% of the sequencing reads reached Q30 (99.9% base call accuracy). RNA-seq and ChIP-seq analysis and pathway enrichment analysis All RNA-seq and ChIP-seq datasets have been deposited in the Gene Expression Ominbus (GEO) with the accession ID ([201]GSE206112). RNA-seq The reads were mapped to the human genome hg38 using STAR (v2.7.2a).[202]^39 RNA-seq aligner with the following parameter: “--outSAMmapqUnique 60”. Uniquely mapped sequencing reads were assigned to Gencode 31 gene using featureCounts (v1.6.2)[203]^40 with the following parameters: "-s 2 –p –Q 10 -O”. The data was filtered using read count >10 in at least 3 of the samples, normalized using TMM (trimmed mean of M values) method and subjected to differential expression analysis using edgeR (v3.36.0).[204]^41^,[205]^42 Gene ontology and pathway functional analysis was performed on differential expression gene with false discovery rate cut-off of 0.05 and absolute value of log2 of fold change cut-off of 0.25 using DAVID.[206]^43^,[207]^44 Gene set enrichment analysis was conducted by using hypergeometric test against human gene ontology and MsigDB v.6 canonical pathways, with p < 0.01 as the significant cutoff.[208]^45 ChIP-seq Bowtie2[209]^46 was used for ChIP-Seq reads alignment on the human genome (hg38). Duplicated reads were removed using Picard [ref. 1]. Peak calling of mapped ChIP-Seq reads were performed by MACS2[210]^47 compared with input ChIP-Seq with a bonferroni adjusted cutoff of p value less than 0.01. Peaks called from multiple samples were merged. Merged peaks overlapping with ENCODE blacklist regions[211]^48^,[212]^49 were removed to form a final set of regions. Reads overlapping with these regions in different samples were counted by featureCounts.[213]^40 The data was filtered using at least 10 read counts in more than one of the samples, normalized using TMM (trimmed mean of M values) method and subjected to differential analysis using edgeR (v3.36.0).[214]^41^,[215]^42 Gene ontology and pathway functional analysis was performed on genes whose upstream 2kbp having differential peak signals with false discovery rate cut-off of 0.05 and absolute value of log2 of fold change cut-off of 0.25 using DAVID.[216]^43 Ref. 1: Broad Institute. (Accessed: 2018/02/21; version 2.17.8). “Picard Tools.” Broad Institute, GitHub repository. [217]http://broadinstitute.github.io/picard/. Quantification and statistical analysis Each experiment was performed in replicates (biological and technical) to confirm observed phenotypes and data reproducibility. Data presented in figures was created in GraphPad Prism 9 and plotted as mean ± S.E.M. Statistical analysis was done in GraphPad Prism 9 using two-sided paired or unpaired t-tests. p values ≤0.05 were considered statistically significant. Acknowledgments