Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A non-canonical tricarboxylic acid cycle underlies cellular identity

Abstract

The tricarboxylic acid (TCA) cycle is a central hub of cellular metabolism, oxidizing nutrients to generate reducing equivalents for energy production and critical metabolites for biosynthetic reactions. Despite the importance of the products of the TCA cycle for cell viability and proliferation, mammalian cells display diversity in TCA-cycle activity1,2. How this diversity is achieved, and whether it is critical for establishing cell fate, remains poorly understood. Here we identify a non-canonical TCA cycle that is required for changes in cell state. Genetic co-essentiality mapping revealed a cluster of genes that is sufficient to compose a biochemical alternative to the canonical TCA cycle, wherein mitochondrially derived citrate exported to the cytoplasm is metabolized by ATP citrate lyase, ultimately regenerating mitochondrial oxaloacetate to complete this non-canonical TCA cycle. Manipulating the expression of ATP citrate lyase or the canonical TCA-cycle enzyme aconitase 2 in mouse myoblasts and embryonic stem cells revealed that changes in the configuration of the TCA cycle accompany cell fate transitions. During exit from pluripotency, embryonic stem cells switch from canonical to non-canonical TCA-cycle metabolism. Accordingly, blocking the non-canonical TCA cycle prevents cells from exiting pluripotency. These results establish a context-dependent alternative to the traditional TCA cycle and reveal that appropriate TCA-cycle engagement is required for changes in cell state.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Genetic co-essentiality mapping of metabolic enzymes reveals two TCA-cycle modules.
Fig. 2: ACL loss disrupts TCA-cycle metabolism in ES cells.
Fig. 3: Engagement of the non-canonical TCA cycle is cell-state dependent.
Fig. 4: Exit from naive pluripotency requires engagement of the non-canonical TCA cycle.

Similar content being viewed by others

Data availability

RNA-seq data supporting the findings of this study have been deposited in the Gene Expression Omnibus under the accession code GSE183434. Alignment was performed against the mouse mm10 genome assembly. Gene essentiality data and NSCLC gene expression data are available from the DepMap Portal (https://depmap.org/portal/). Isotopologue distributions from all MS isotope tracing experiments are provided in Supplementary Table 4Source data are provided with this paper.

Code availability

The code used to perform gene essentiality correlation and network modelling is provided at GitHub (https://github.com/finley-lab/coessentiality-network).

References

  1. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  2. Kim, J. & DeBerardinis, R. J. Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab. 30, 434–446 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Muir, A., Danai, L. V. & Vander Heiden, M. G. Microenvironmental regulation of cancer cell metabolism: implications for experimental design and translational studies. Dis. Model. Mech. 11, dmm035758 (2018).

    Article  Google Scholar 

  4. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wainberg, M. et al. A genome-wide atlas of co-essential modules assigns function to uncharacterized genes. Nat. Genet. 53, 638–649 (2021).

  6. Hatzivassiliou, G. et al. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 8, 311–321 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Zhao, S. et al. ATP-citrate lyase controls a glucose-to-acetate metabolic switch. Cell Rep. 17, 1037–1052 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chen, P. H. et al. Metabolic diversity in human non-small cell lung cancer cells. Mol. Cell 76, 838–851 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Alves, T. C. et al. Integrated, step-wise, mass-isotopomeric flux analysis of the TCA cycle. Cell Metab. 22, 936–947 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sullivan, L. B. et al. Aspartate is an endogenous metabolic limitation for tumour growth. Nat. Cell Biol. 20, 782–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lewis, C. A. et al. Tracing compartmentalized NADPH metabolism in the cytosol and mitochondria of mammalian cells. Mol. Cell 55, 253–263 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Williamson, D. H., Lund, P. & Krebs, H. A. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem. J. 103, 514–527 (1967).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Krebs, H. A. & Eggleston, L. V. The oxidation of pyruvate in pigeon breast muscle. Biochem. J. 34, 442–459 (1940).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shintaku, J. et al. MyoD regulates skeletal muscle oxidative metabolism cooperatively with alternative NF-κB. Cell Rep. 17, 514–526 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Stacpoole, P. W. The pharmacology of dichloroacetate. Metabolism 38, 1124–1144 (1989).

    Article  CAS  PubMed  Google Scholar 

  17. Kalkan, T. et al. Tracking the embryonic stem cell transition from ground state pluripotency. Development 144, 1221–1234 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Vardhana, S. A. et al. Glutamine independence is a selectable feature of pluripotent stem cells. Nat. Metab. 1, 676–687 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Pereira, L., Yi, F. & Merrill, B. J. Repression of Nanog gene transcription by Tcf3 limits embryonic stem cell self-renewal. Mol. Cell. Biol. 26, 7479–7491 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wray, J. et al. Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat. Cell Biol. 13, 838–845 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Moussaieff, A. et al. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 21, 392–402 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Borst, P. The malate-aspartate shuttle (Borst cycle): how it started and developed into a major metabolic pathway. IUBMB Life 72, 2241–2259 (2020).

  24. Assmann, N. et al. Srebp-controlled glucose metabolism is essential for NK cell functional responses. Nat. Immunol. 18, 1197–1206 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Luengo, A. et al. Increased demand for NAD+ relative to ATP drives aerobic glycolysis. Mol. Cell 81, 691–707 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Davidson, S. M. et al. Environment impacts the metabolic dependencies of Ras-driven non-small cell lung cancer. Cell Metab. 23, 517–528 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Marin-Valencia, I. et al. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 15, 827–837 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhu, X. G. et al. Functional genomics in vivo reveal metabolic dependencies of pancreatic cancer cells. Cell Metab. 33, 211–221 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Dempster, J. M. et al. Extracting biological insights from the project achilles genome-scale CRISPR screens in cancer cell lines. Preprint at https://doi.org/10.1101/720243 (2019).

  30. Meyers, R. M. et al. Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells. Nat. Genet. 49, 1779–1784 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ashburner, M. et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. The Gene Ontology Consortium. The Gene Ontology Resource: 20 years and still GOing strong. Nucleic Acids Res. 47, D330–D338 (2019).

    Article  Google Scholar 

  33. Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods 17, 261–272 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ghandi, M. et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508 (2019).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  35. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  36. Carey, B. W., Finley, L. W., Cross, J. R., Allis, C. D. & Thompson, C. B. Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 518, 413–416 (2015).

    Article  CAS  PubMed  ADS  Google Scholar 

  37. Dow, L. E. et al. Inducible in vivo genome editing with CRISPR-Cas9. Nat. Biotechnol. 33, 390–394 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. MacDougall, M. S., Clarke, R. & Merrill, B. J. Intracellular Ca2+ homeostasis and nuclear export mediate exit from naive pluripotency. Cell Stem Cell 25, 210–224 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Morris, J. P. T. et al. α-Ketoglutarate links p53 to cell fate during tumour suppression. Nature 573, 595–599 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Fellmann, C. et al. An optimized microRNA backbone for effective single-copy RNAi. Cell Rep. 5, 1704–1713 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Millard, P. et al. IsoCor: isotope correction for high-resolution MS labeling experiments. Bioinformatics 35, 4484–4487 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the members of the Finley laboratory for discussion, and A. Boire, A. Intlekofer, S. Vardhana, E. Reznik and K. S. Tan for feedback. Rex1::GFPd2 cells were a gift from A. Smith. P.K.A. is an NICHD Ruth L. Kirschstein Predoctoral fellow (F31HD098824). B.T.J. is a Gerstner Sloan Kettering Grayer Fellow and is supported by a Medical Scientist Training Program grant from the NIGMS of the National Institutes of Health under award number T32GM007739 to the Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program. K.I.P. is supported by a Bruce Charles Forbes Pre-Doctoral Fellowship (MSKCC). J.S.B. is supported by a Human Frontier Science Program Fellowship (LT000200/2021-L). L.B.S. is supported by R00CA218679 and P30CA015704. L.W.S.F. is a Searle Scholar. This research was also supported by grants to L.W.S.F. from the Pershing Square Sohn Prize for Cancer Research, the Starr Foundation (I12-0051), the NIH/NCI (R37 CA252305) and the Anna Fuller Fund as well as the Memorial Sloan Kettering Cancer Center Support Grant P30CA008748.

Author information

Authors and Affiliations

Authors

Contributions

P.K.A. and L.W.S.F. conceived the study. P.K.A. and B.T.J. performed all of the experiments with assistance from K.I.P., J.S.B. and J.E.; M.L.H., O.J.N. and S.P.A. performed LC–MS experiments under the guidance of L.B.S.; B.T.J. performed genetic co-essentiality mapping and network modelling with guidance from E.D. K.I.P. performed RNA-seq. L.B.S. provided additional study guidance. L.W.S.F. supervised the project. P.K.A. and L.W.S.F. wrote the manuscript with input from all of the authors.

Corresponding author

Correspondence to Lydia W. S. Finley.

Ethics declarations

Competing interests

P.K.A., B.T.J. and L.W.S.F. are listed as inventors on a provisional patent application (US provisional application no. 63/272,940) filed by the Memorial Sloan Kettering Cancer Center. The patent application covers the use of ACL inhibitors to modify the self-renewal potential of ES cells. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Metabolic gene essentiality correlations across cancer cell lines.

Heatmap depicting hierarchical clustering of pairwise gene essentiality score correlations of core metabolic pathway genes derived from four GO terms: tricarboxylic acid (TCA) cycle, canonical glycolysis, one-carbon metabolic process and fatty-acyl-CoA metabolic process. Genes are colour coded to the left of the heatmap according to the GO term. TCA cycle genes are highlighted (red) in the dendrogram. Gene names and correlation scores can be found in Supplementary Table 1.

Extended Data Fig. 2 Effect of ACL inhibition on 13C labelling of TCA cycle metabolites.

a, Two-dimensional network diagram representing gene essentiality score correlations between TCA cycle genes and their top co-dependencies. The strength of the correlation between genes is represented by both the length and thickness of the connecting edge. Correlation scores are shown in Supplementary Table 1. b, c, Fractional enrichment of citrate (left) and malate (right) in three NSCLC cell lines cultured in medium containing [U-13C]glucose (b) or [U-13C]glutamine (c) and treated with vehicle or 50 μM BMS-303141(ACLi) for 24 h. d, Schematic depicting [U-13C]asparagine labelling of aspartate and citrate in cells expressing guinea pig asparaginase (ASNase). Asparagine-derived aspartate will generate M+4-labelled citrate. Top, M+4-labelled citrate metabolized via the canonical TCA cycle will lose two labelled carbons as CO2, ultimately regenerating citrate that retains two labelled carbons (M+2). Bottom, M+4-labelled citrate metabolized by ACL will yield M+4 labelled oxaloacetate that will ultimately regenerate M+4-labelled citrate. e, f, Fractional labelling of aspartate (left) and citrate (right) (e) or citrate M+2 relative to citrate M+4 (cit+2/cit+4) (f) in ASNase-expressing 143B human osteosarcoma cells cultured in medium containing [U-13C]asparagine and treated with vehicle or 50 μM ACLi for 24 h. Data are mean ± SD, n = 3 independent replicates. Significance was assessed in comparison to vehicle treatment by two-way ANOVA with Sidak’s multiple comparisons post-test (b-c, e) or using unpaired two-tailed Student’s t-test (f).

Source data

Extended Data Fig. 3 ACO2 and ACL disruption in embryonic stem cells.

a, Fractional M+2 enrichment of citrate and malate in mouse ES cells cultured in medium containing [U-13C]glucose. b, Fractional enrichment of malate M+2 relative to citrate M+2 (mal+2/cit+2) derived from [U-13C]glucose in ES cells following treatment with vehicle or 50 μM BMS-303141 (ACLi) for 24 h. c, d, Immunoblot of clonal mouse ES cells in which CRISPR/Cas9-mediated editing was used to target either a non-genic region of chromosome 8 (Ctrl) and Acly (ACLY-1 and ACLY-2) (c) or Aco2 (ACO2-1 and ACO2-2) (d). e, f, Assessment of the [U-13C]glucose-derived mal+2/cit+2 ratio (e) or steady-state levels of TCA cycle metabolites represented as the fold change (expressed in log2) relative to Ctrl (f) in control and Aco2-edited ES cells. g, h, j, k, Fractional M+1 enrichment of NADH (g), lactate (h), fumarate (j) and succinate (k) in control and Acly-edited ES cells cultured in medium containing [4-2H]glucose. i, Schematic depicting deuterium transfer from [4-2H]glucose first onto malate in the cytoplasm then onto TCA cycle metabolites in the mitochondria. l, Quantification of the lactate over pyruvate ratio in control and Acly-edited ES cells. m, The baseline oxygen consumption rate (OCR) in control and Acly-edited ES cells normalized to protein content. Twelve technical replicates were averaged for each of three independent experiments. Data are mean ± SD, n = 3 independent replicates unless otherwise noted. Significance was assessed using unpaired two-tailed Student’s t-test (a, b) or in comparison to control cells by one-way ANOVA with Sidak’s multiple comparisons post-test for all other panels.

Source data

Extended Data Fig. 4 SLC25A1 and MDH1 contribute to TCA cycle metabolism in embryonic stem cells.

a, b, Immunoblot of clonal mouse ES cells in which CRISPR/Cas9-mediated editing was used to target either a non-genic region of chromosome 8 (Ctrl) and Slc25a1 (SLC25A1-1 and SLC25A1-2) (a) or Mdh1 (MDH1-1 and MDH1-2) (b). c, d, Fractional M+1 enrichment of malate (Mal), fumarate (Fum), aspartate (Asp) and citrate (Cit) in control (Ctrl) and Slc25a1-edited ES cells (c) or Mdh1-edited ES cells (d) cultured in medium containing [4-2H]glucose. e, f, Fractional M+2 enrichment of citrate, fumarate, malate and aspartate derived from [U-13C]glucose in control and Slc25a1-edited (e) or Mdh1-edited (f) ES cells. g, Steady-state levels of TCA cycle metabolites in Slc25a1-edited or Mdh1-edited ES cells. Levels are represented as the fold change (expressed in log2) relative to chromosome 8-targeted control cells. Data are mean ± SD, n = 3 independent replicates. Significance was assessed in comparison to control cells by two-way ANOVA (c-f) with Sidak’s multiple comparisons post-test.

Source data

Extended Data Fig. 5 Effect of myogenic differentiation on 13C-glucose labelling of TCA cycle intermediates.

a, Immunoblot comparing expression of myogenesis markers MYOG and MYH3 between proliferating (Prolif) and 100% confluent (Conf) myoblasts and myotubes differentiated for 3, 5 or 7 days. b, Fractional labelling of citrate (left) and malate (right) in proliferating and confluent myoblasts and myotubes differentiated for 3, 5 or 7 days cultured in medium containing [U-13C]glucose. c, Fractional M+1 enrichment from [4-2H]glucose of malate (Mal), fumarate (Fum), aspartate (Asp) and citrate (Cit) in myoblasts and myotubes differentiated for 5 days. d, Immunoblot comparing expression of ACL and ACO2 in C2C12 cells expressing doxycycline-inducible shRNAs targeting Acly (shAcly-1 and shAcly-2), Aco2 (shAco2-1 and shAco2-2) or Renilla luciferase (shRen, used as a control). Cells were cultured on doxycycline for two days to induce shRNA expression. e-h, Fractional M+2 enrichment of citrate (left) and malate (right) or malate M+2 relative to citrate M+2 (mal+2/cit+2) in myoblasts (e, f) or myotubes (g, h) expressing doxycycline-inducible shRNAs targeting Acly, Aco2 or Renilla luciferase cultured in medium containing [U-13C]glucose. Myoblasts and myotubes were cultured on doxycycline for 2 or 4 days, respectively, to induce shRNA expression. Data are mean ± SD, n = 3 independent replicates. In b, significance was assessed using one-way ANOVA with Sidak’s multiple comparisons post-test to compare total metabolite fraction labelled relative to proliferating myoblasts. In remaining panels, significance was assessed by two-way ANOVA in comparison to myoblasts (c) or by one-way ANOVA in comparison to shRen-expressing myoblasts (e-f) or myotubes (g-h) with Sidak’s multiple comparisons post-test.

Source data

Extended Data Fig. 6 Transcriptional profiles associated with TCA cycle choice.

a, Gene set enrichment analysis showing that genes positively correlated with fractional enrichment of malate M+2 relative to citrate M+2 (mal+2/cit+2) derived from [U-13C]glucose in 68 NSCLC cell lines are enriched for KEGG citric acid (TCA) cycle-associated genes. b, c, Fractional M+2 enrichment of citrate (Cit), fumarate (Fum), malate (Mal) and aspartate (Asp) (b) or mal+2/cit+2 (c) derived from [U-13C]glucose in mouse ES cells following treatment with vehicle, 5 mM DCA or 10 µM MPCi for 24 h. Data are mean ± SD, n = 3 independent samples. In b-c, significance was assessed in comparison to vehicle treatment by two-way ANOVA (b) or one-way ANOVA (c) with Sidak’s multiple comparisons post-test.

Source data

Extended Data Fig. 7 ACL loss blunts exit from naive pluripotency.

a, Experimental setup for cell fate transitions. Mouse ES cells cultured in serum and leukemia inhibitory factor (LIF) are a heterogenous population that can be converted to the naive, ground state of pluripotency by addition of MEK and GSK3β inhibitors (2i) in either serum replete (serum/LIF+2i, d-f) or serum-free (2i/LIF, g-i) media formulations. Transition to serum-free medium lacking 2i/LIF (−2i/LIF) induces exit from the naive pluripotent state, enabling ES cells to gain differentiation competence. b, RT–qPCR of pluripotency-associated (Nanog, Esrrb, Klf2, Rex1 and Oct4) and early differentiation-associated (Fgf5, Otx2 and Sox1) genes in 2i/LIF-cultured ES cells subjected to 2i/LIF withdrawal for 12, 24 or 40 h. Levels are represented as the fold change (expressed in log2) relative to naive, 2i/LIF-cultured ES cells (0 h). c, Quantification of alkaline phosphatase (AP)-positive colonies representing ES cells that failed to exit from the pluripotent state. 2i/LIF-cultured ES cells were subjected to 2i/LIF withdrawal for 0, 12, 24 or 40 h and then reseeded at clonal density into medium containing 2i and LIF. d-f, Fractional labelling of citrate (Cit), malate (Mal) and aspartate (Asp) in serum/LIF+2i-cultured ES cells incubated with [U-13C]glucose (d), [U-13C]glutamine (e) or [4-2H]glucose (f) subjected to exit from pluripotency for the indicated times. g, Fractional enrichment of glucose-derived malate M+2 relative to citrate M+2 (mal+2/cit+2) in 2i/LIF-cultured ES cells subjected to 2i/LIF withdrawal for the indicated times. h, i, Fractional labelling of citrate, malate and aspartate in 2i/LIF-cultured ES cells cultured in medium containing [U-13C]glucose (h) or [U-13C]glutamine (i) subjected to 2i/LIF withdrawal for the indicated times. j, Immunoblot of polyclonal ES cells in which CRISPR/Cas9-mediated editing was used to target either a non-genic region of chromosome 8 (sgChr8) or Tcf7l1 (sgTcf7l1). k, RT–qPCR of pluripotency-associated (Nanog,Esrrb, and Rex1) and early differentiation-associated (Sox1) genes in control and Tcf7l1-edited ES cells adapted to the naive, ground state and subjected to 2i/LIF withdrawal for the indicated times. Levels are represented as the fold change (expressed in log2) relative to chromosome 8-targeted control cells in the naive, ground state (0 h). l, m, Fractional labelling of citrate (left) and malate (right) (l) and glucose-derived mal+2/cit+2 ratio (m) in chromosome 8-targeted control or Tcf7l1-edited ES cells cultured in medium containing [U-13C]glucose subjected to 2i/LIF withdrawal for the indicated times. Data are mean ± SD, n = 3 independent replicates. In d-e, h-i, and l, significance was assessed using one-way ANOVA (d-e, h-i) or two-way ANOVA (l) with Sidak’s multiple comparisons post-test to compare total metabolite fraction labelled relative to the 0 h timepoint (d-e, h-i) or control cells (l). In remaining panels, significance was assessed relative to the 0 h timepoint using one-way ANOVA (c, f-g) or chromosome 8-targeted control cells at each time point using two-way ANOVA (k, m) with Sidak’s multiple comparisons post-test.

Source data

Extended Data Fig. 8 Acetate does not reverse the effects of ACL loss on exit from pluripotency.

a, b, Fractional labelling of citrate (Cit), malate (Mal) and aspartate (Asp) in control and Acly-edited ES cells cultured in medium containing [U-13C]glucose (a) or [U-13C]glutamine (b) following 40 h of 2i/LIF withdrawal. c, d, Fractional enrichment of malate M+2 relative to citrate M+2 (mal+2/cit+2) derived from [U-13C]glucose (c) or steady-state levels of TCA cycle metabolites (d) in naive, 2i-adapted control (Ctrl) and Acly-edited (ACLY-1 and ACLY-2) ES cells. Steady-state levels are represented as the fold change (expressed in log2) relative to control cells. e, f, Assessment of the [U-13C]glucose-derived mal+2/cit+2 ratio (e) and steady-state levels of TCA cycle metabolites (f) in control and Acly-edited ES cells subjected to 2i/LIF withdrawal for 40 h. g, Relative viability (measured by PI exclusion) of control and Acly-edited ES cells maintained in the naive pluripotent state (+2i/LIF, left) or subjected to 2i/LIF withdrawal for 40 h (−2i/LIF, right). h, Immunoblot showing expression of ACSS2, the enzyme that converts acetate to acetyl-CoA in the cytosol, in naive, ground state ES cells subjected to 2i/LIF withdrawal for the indicated times. i, Fractional labelling of palmitate in control and Acly-edited ES cells cultured in medium containing [U-13C]acetate following 40 h of 2i/LIF withdrawal. Each bar represents one independent sample. j, Immunoblot comparing levels of acetylation (ac) at indicated histone lysine residues in control and Acly-edited ES cells subjected to 2i/LIF withdrawal for 40 h in the presence of vehicle or 5 mM sodium acetate. k, Relative viability of control and Acly-edited ES cells subjected to 2i/LIF withdrawal for 40 h in the presence of vehicle or 5 mM sodium acetate. l, Quantification of GFP mean fluorescence intensity (MFI) encoded by the Rex1::GFPd2 reporter in ES cells subjected to 2i/LIF withdrawal for 40 h in the presence of vehicle or 50 μM BMS-303141 (ACLi). Naïve ES cells (+2i/LIF) are included as a control. Representative histograms are shown in Fig. 4d. m, RT–qPCR of pluripotency-associated (Nanog, Esrrb and Rex1) and early differentiation-associated (Sox1) genes in control and Acly-edited ES cells subjected to 2i/LIF withdrawal for 40 h. Levels are represented as the fold change (expressed in log2) relative to chromosome 8-targeted control cells. n, Alkaline phosphatase (AP) staining of colony formation assay representing control and Acly-edited ES cells that failed to exit the naive pluripotent state. 2i-adapted ES cells were subjected to 2i/LIF withdrawal for 40 h and then reseeded at clonal density into medium containing 2i/LIF. Quantification is shown in Fig. 4e. o, RT–qPCR of pluripotency-associated genes in control and Acly-edited ES cells subjected to 2i/LIF withdrawal for 40 h in the presence of vehicle or 5 mM sodium acetate. p, Quantification of GFP MFI encoded by the Rex1::GFPd2 reporter in ES cells subjected to 2i/LIF withdrawal for 40 h in the presence of DMSO or 50 μM BMS-303141(ACLi) and vehicle or 5 mM sodium acetate. q, Quantification of AP-positive colonies representing control and Acly-edited ES cells that failed to exit from the pluripotent state. ES cells were subjected to 2i/LIF withdrawal for 40 h in the presence of vehicle or 5 mM sodium acetate prior to reseeding at clonal density into medium containing 2i and LIF. Data are mean ± SD, n = 5 (p), n = 4 (g, k, l) or n = 3 (all other experiments) independent replicates. In a-b, significance was assessed using one-way ANOVA with Sidak’s multiple comparisons post-test to compare total metabolite fraction labelled relative to control cells. In remaining panels, significance was assessed by two-way ANOVA relative to control cells (k, q) or DMSO treatment (p) with Sidak’s multiple comparisons post-test, or by one-way ANOVA in comparison to control cells (c, e, g) with Sidak’s multiple comparisons post-test or in the indicated comparisons (l) with Tukey’s multiple comparisons post-test.

Source data

Extended Data Fig. 9 Effect of SLC25A1 and MDH1 loss in exit from naive pluripotency.

a, b, Relative viability (measured by PI exclusion) of control and Slc25a1-edited (left) and Mdh1-edited (right) ES cells maintained in the naive pluripotent state (+2i/LIF, a) or subjected to 2i/LIF withdrawal for 40 h (−2i/LIF, b). c, Steady-state levels of TCA cycle metabolites in control and Slc25a1-edited (left) and Mdh1-edited (right) ES cells subjected to 2i/LIF withdrawal for 40 h. Steady-state levels are represented as the fold change (expressed in log2) relative to control cells. d, e, Relative O-propargyl-puromycin (OP-puro) mean fluorescence intensity (MFI) in control, Slc25a1-edited, Acly-edited and Mdh1-edited ES cells that have been maintained in the naive pluripotent state (d) or subjected to 2i/LIF withdrawal for 40 h (e). Dotted line represents OP-puro MFI following cycloheximide (CHX) treatment as a control. f, g, Population doublings of control, Slc25a1-edited, Acly-edited and Mdh1-edited ES cells that have been maintained in the naive pluripotent state (f) or subjected to 2i/LIF withdrawal for 40 h (g). h, i, RT–qPCR of pluripotency-associated (Nanog, Esrrb and Rex1) and early differentiation-associated (Sox1) genes in control and Slc25a1-edited (h) and Mdh1-edited (i) ES cells subjected to 2i/LIF withdrawal for 40 h. Data are mean ± SD, n = 3 independent samples. Significance was assessed in comparison to control cells by one-way ANOVA with Sidak’s multiple comparisons post-test.

Source data

Extended Data Fig. 10 Mode of TCA cycle engagement regulates cell fate.

a, Population doublings of control and Aco2-edited ES cells cultured in metastable (serum/LIF) conditions. b, Cumulative population doublings over the indicated passages of control and Aco2-edited ES cells upon conversion to the naive, ground state of pluripotency via addition of MEK and GSK3β inhibitors (+2i). c, RT–qPCR of pluripotency-associated genes at the indicated passages in control and Aco2-edited ES cells following addition of 2i. Gene expression at every passage was normalized to passage 0 (p0). Data are mean ± SD, n = 1 (b) or n = 3 (a, c) independent replicates. Significance was assessed in comparison to control cells by one-way ANOVA with Sidak’s multiple comparisons post-test (a) or relative to control cells at each timepoint with P values coloured according to comparison by two-way ANOVA with Sidak’s multiple comparisons post-test (c).

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1: uncropped scans of source data for immunoblots. Supplementary Fig. 2: gating strategy for FACS analysis.

Reporting Summary

Peer Review File

Supplementary Table 1

Source data for Fig. 1a and Extended Data Figs. 1 and 2a. Correlations derived from gene co-essentiality analysis. Data underlie the 2D network diagrams shown in Fig. 1a and Extended Data Fig. 2a. and the heat map shown in Extended Data Fig. 1.

Supplementary Table 2

All sgRNA, shRNA and RT–qPCR primer sequences. Primer sequences for sgRNAs used to perform CRISPR–Cas9-mediated editing in mouse ES cells to generate clonal lines, shRNAs used to infect C2C12 mouse cells and gene expression analysis using RT–qPCR.

Supplementary Table 3

CRISPR-sequencing primers and derived amplicons. The primer sequences used to amplify genomic fragments containing edited loci of interest for sgRNA editing analysis in clonal mouse ES cell lines and the associated amplicons derived from this analysis. Amplicons are annotated to show editing of each allele for all clonal ES cell lines.

Supplementary Table 4

Source data for all isotope tracing MS experiments. Full isotopologue distributions generated by isotope-tracing MS experiments.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Arnold, P.K., Jackson, B.T., Paras, K.I. et al. A non-canonical tricarboxylic acid cycle underlies cellular identity. Nature 603, 477–481 (2022). https://doi.org/10.1038/s41586-022-04475-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04475-w

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer