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:

Allosteric regulation of CAD modulates de novo pyrimidine synthesis during the cell cycle

Abstract

Metabolism is a fundamental cellular process that is coordinated with cell cycle progression. Despite this association, a mechanistic understanding of cell cycle phase-dependent metabolic pathway regulation remains elusive. Here we report the mechanism by which human de novo pyrimidine biosynthesis is allosterically regulated during the cell cycle. Combining traditional synchronization methods and metabolomics, we characterize metabolites by their accumulation pattern during cell cycle phases and identify cell cycle phase-dependent regulation of carbamoyl-phosphate synthetase 2, aspartate transcarbamylase and dihydroorotase (CAD), the first, rate-limiting enzyme in de novo pyrimidine biosynthesis. Through systematic mutational scanning and structural modelling, we find allostery as a major regulatory mechanism that controls the activity change of CAD during the cell cycle. Specifically, we report evidence of two Animalia-specific loops in the CAD allosteric domain that involve sensing and binding of uridine 5′-triphosphate, a CAD allosteric inhibitor. Based on homology with a mitochondrial carbamoyl-phosphate synthetase homologue, we identify a critical role for a signal transmission loop in regulating the formation of a substrate channel, thereby controlling CAD activity.

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: Cell cycle metabolomics reveals cell cycle-dependent de novo pyrimidine biosynthesis through modulating CAD activity.
Fig. 2: Regulation of cell cycle-dependent CAD activity is independent of mechanisms previously identified.
Fig. 3: Cell cycle-dependent CAD activation is allosterically regulated by UTP binding through two Animalia-specific loops of CAD allosteric domain.
Fig. 4: UTP binding in CAD allosteric domain can alter substrate channel formation through allosteric loop-mediated T′-loop stabilization.
Fig. 5: Proposed mechanism of cell cycle-dependent de novo pyrimidine biosynthesis.

Similar content being viewed by others

Data availability

Publicly available data used in this study include PDB 5DOU, 5DOT, 5G1O and 4C6D. Source data are provided with this paper. All other data are available in the main text or the Supplementary Information.

References

  1. Morgan, D. O. The Cell Cycle: Principles of Control: Primers in Biology (New Science Press, 2007).

  2. D’Angiolella, V. et al. Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair. Cell 149, 1023–1034 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Pavlova, N. N., Zhu, J. & Thompson, C. B. The hallmarks of cancer metabolism: still emerging. Cell Metab. 34, 355–377 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. Ginzberg, M. B. et al. Cell size sensing in animal cells coordinates anabolic growth rates and cell cycle progression to maintain cell size uniformity. Elife 7, e26957 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Del Cano-Ochoa, F., Moreno-Morcillo, M. & Ramon-Maiques, S. CAD, a multienzymatic protein at the head of de novo pyrimidine biosynthesis. Subcell. Biochem. 93, 505–538 (2019).

    Article  PubMed  Google Scholar 

  6. Sigoillot, F. D., Berkowski, J. A., Sigoillot, S. M., Kotsis, D. H. & Guy, H. I. Cell cycle-dependent regulation of pyrimidine biosynthesis. J. Biol. Chem. 278, 3403–3409 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Morford, G., Davidson, J. N. & Snow, E. C. Appearance of CAD activity, the rate-limiting enzyme for pyrimidine biosynthesis, as B cells progress into and through the G1 stage of the cell cycle. Cell Immunol. 158, 96–104 (1994).

    Article  CAS  PubMed  Google Scholar 

  8. Sigoillot, F. D., Sigoillot, S. M. & Guy, H. I. Breakdown of the regulatory control of pyrimidine biosynthesis in human breast cancer cells. Int. J. Cancer 109, 491–498 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Sigoillot, F. D., Evans, D. R. & Guy, H. I. Growth-dependent regulation of mammalian pyrimidine biosynthesis by the protein kinase A and MAPK signaling cascades. J. Biol. Chem. 277, 15745–15751 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Musmanno, L. A., Jamison, R. S., Barnett, R. S., Buford, E. & Davidson, J. N. Complete hamster CAD protein and the carbamylphosphate synthetase domain of CAD complement mammalian cell mutants defective in de novo pyrimidine biosynthesis. Somat. Cell Mol. Genet. 18, 309–318 (1992).

    Article  CAS  PubMed  Google Scholar 

  12. Swyryd, E. A., Seaver, S. S. & Stark, G. R. N-(phosphonacetyl)-l-aspartate, a potent transition state analog inhibitor of aspartate transcarbamylase, blocks proliferation of mammalian cells in culture. J. Biol. Chem. 249, 6945–6950 (1974).

    Article  CAS  PubMed  Google Scholar 

  13. Agarwal, M. L. et al. A p53-dependent S-phase checkpoint helps to protect cells from DNA damage in response to starvation for pyrimidine nucleotides. Proc. Natl Acad. Sci. USA 95, 14775–14780 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ruhul Amin, A. R. et al. N-(phosphonacetyl)-l-aspartate induces TAp73-dependent apoptosis by modulating multiple Bcl-2 proteins: potential for cancer therapy. Oncogene 32, 920–929 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Del Cano-Ochoa, F. et al. Cell-based analysis of CAD variants identifies individuals likely to benefit from uridine therapy. Genet. Med. 22, 1598–1605 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Miltenberger, R. J., Sukow, K. A. & Farnham, P. J. An E-box-mediated increase in cad transcription at the G1/S-phase boundary is suppressed by inhibitory c-Myc mutants. Mol. Cell. Biol. 15, 2527–2535 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Graves, L. M. et al. Regulation of carbamoyl phosphate synthetase by MAP kinase. Nature 403, 328–332 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Sigoillot, F. D. et al. Protein kinase C modulates the up-regulation of the pyrimidine biosynthetic complex, CAD, by MAP kinase. Front. Biosci. 12, 3892–3898 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Banerjei, L. C. & Davidson, J. N. Site-directed substitution of Ser1406 of hamster CAD with glutamic acid alters allosteric regulation of carbamyl phosphate synthetase II. Somat. Cell Mol. Genet. 23, 37–49 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Robitaille, A. M. et al. Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis. Science 339, 1320–1323 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Ben-Sahra, I., Howell, J. J., Asara, J. M. & Manning, B. D. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 339, 1323–1328 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tatibana, M. & Shigesada, K. Activation by 5-phosphoribosyl 1-pyrophosphate of glutamine-dependent carbamyl phosphate synthetase from mouse spleen. Biochem. Biophys. Res. Commun. 46, 491–497 (1972).

    Article  CAS  PubMed  Google Scholar 

  23. Tatibana, M. & Shigesada, K. Control of pyrimidine biosynthesis in mammalian tissues. V. Regulation of glutamine-dependent carbamyl phosphate synthetase: activation by 5-phosphoribosyl 1-pyrophosphate and inhibition by uridine triphosphate. J. Biochem. 72, 549–560 (1972).

    Article  CAS  PubMed  Google Scholar 

  24. Santos, A., Wernersson, R. & Jensen, L. J. Cyclebase 3.0: a multi-organism database on cell-cycle regulation and phenotypes. Nucleic Acids Res. 43, D1140–D1144 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Lee, L., Kelly, R. E., Pastra-Landis, S. C. & Evans, D. R. Oligomeric structure of the multifunctional protein CAD that initiates pyrimidine biosynthesis in mammalian cells. Proc. Natl Acad. Sci. USA 82, 6802–6806 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Moreno-Morcillo, M. et al. Structural insight into the Core of CAD, the multifunctional protein leading de novo pyrimidine biosynthesis. Structure 25, 912–923 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Ruiz-Ramos, A., Velazquez-Campoy, A., Grande-Garcia, A., Moreno-Morcillo, M. & Ramon-Maiques, S. Structure and functional characterization of human aspartate transcarbamoylase, the target of the anti-tumoral drug PALA. Structure 24, 1081–1094 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Qiu, Y. & Davidson, J. N. Substitutions in the aspartate transcarbamoylase domain of hamster CAD disrupt oligomeric structure. Proc. Natl Acad. Sci. USA 97, 97–102 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Traut, T. W. Physiological concentrations of purines and pyrimidines. Mol. Cell. Biochem. 140, 1–22 (1994).

    Article  CAS  PubMed  Google Scholar 

  30. Liu, X., Guy, H. I. & Evans, D. R. Identification of the regulatory domain of the mammalian multifunctional protein CAD by the construction of an Escherichia coli hamster hybrid carbamyl-phosphate synthetase. J. Biol. Chem. 269, 27747–27755 (1994).

    Article  CAS  PubMed  Google Scholar 

  31. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protoc. 10, 845–858 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Thoden, J. B., Huang, X., Kim, J., Raushel, F. M. & Holden, H. M. Long-range allosteric transitions in carbamoyl phosphate synthetase. Protein Sci. 13, 2398–2405 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Meister, A. Mechanism and regulation of the glutamine-dependent carbamyl phosphate synthetase of Escherichia coli. Adv. Enzymol. Relat. Areas Mol. Biol. 62, 315–374 (1989).

    CAS  PubMed  Google Scholar 

  34. Cohen, P. P., Hall, L. M. & Metzenberg, R. L. Isolation and characterization of a naturally occurring stimulator of citrulline biosynthesis. Nature 178, 1468–1469 (1956).

    Article  CAS  PubMed  Google Scholar 

  35. Rubio, V., Ramponi, G. & Grisolia, S. Carbamoyl phosphate synthetase I of human liver. Purification, some properties and immunological cross-reactivity with the rat liver enzyme. Biochim. Biophys. Acta 659, 150–160 (1981).

    Article  CAS  PubMed  Google Scholar 

  36. Fresquet, V. et al. Site-directed mutagenesis of the regulatory domain of Escherichia coli carbamoyl phosphate synthetase identifies crucial residues for allosteric regulation and for transduction of the regulatory signals. J. Mol. Biol. 299, 979–991 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Cervera, J. et al. Photoaffinity labeling with UMP of lysine 992 of carbamyl phosphate synthetase from Escherichia coli allows identification of the binding site for the pyrimidine inhibitor. Biochemistry 35, 7247–7255 (1996).

    Article  CAS  PubMed  Google Scholar 

  38. Betts, M. J. & Russell, R. B. Amino acid properties and consequences of substitutions. in Bioinformatics for Geneticists (eds. Barnes, M. R. & Gray, I. C.) 311–342 (2007).

  39. Jafari, R. et al. The cellular thermal shift assay for evaluating drug target interactions in cells. Nat. Protoc. 9, 2100–2122 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. de Cima, S. et al. Structure of human carbamoyl phosphate synthetase: deciphering the on/off switch of human ureagenesis. Sci. Rep. 5, 16950 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Abagyan, R. et al. Homology modeling with internal coordinate mechanics: deformation zone mapping and improvements of models via conformational search. Proteins 1, 29–37 (1997).

    Article  Google Scholar 

  42. Abagyan, R. A. & Totrov, M. M. Contact area difference (CAD): a robust measure to evaluate accuracy of protein models. J. Mol. Biol. 268, 678–685 (1997).

    Article  CAS  PubMed  Google Scholar 

  43. Cardozo, T., Totrov, M. & Abagyan, R. Homology modeling by the ICM method. Proteins 23, 403–414 (1995).

    Article  CAS  PubMed  Google Scholar 

  44. Abagyan, R. & Totrov, M. Biased probability Monte Carlo conformational searches and electrostatic calculations for peptides and proteins. J. Mol. Biol. 235, 983–1002 (1994).

    Article  CAS  PubMed  Google Scholar 

  45. Waterhouse, A. M., Procter, J. B., Martin, D. M., Clamp, M. & Barton, G. J. Jalview Version 2—a multiple sequence alignment editor and analysis workbench. Bioinformatics 25, 1189–1191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. UniProt, C. UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Res. 49, D480–D489 (2021).

    Article  Google Scholar 

  47. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Henikoff, S. & Henikoff, J. G. Amino acid substitution matrices from protein blocks. Proc. Natl Acad. Sci. USA 89, 10915–10919 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Abagyan, R. A. & Batalov, S. Do aligned sequences share the same fold? J. Mol. Biol. 273, 355–368 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Schapira, M. et al. Discovery of diverse thyroid hormone receptor antagonists by high-throughput docking. Proc. Natl Acad. Sci. USA 100, 7354–7359 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Neves, M. A., Totrov, M. & Abagyan, R. Docking and scoring with ICM: the benchmarking results and strategies for improvement. J. Comput. Aided Mol. Des. 26, 675–686 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Arnautova, Y. A., Abagyan, R. A. & Totrov, M. Development of a new physics-based internal coordinate mechanics force field and its application to protein loop modeling. Proteins 79, 477–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sehnal, D. et al. MOLE 2.0: advanced approach for analysis of biomacromolecular channels. J. Cheminform. 5, 39 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We sincerely appreciate E. Freinkman for cell cycle LC–MS metabolomics analysis, Q. Spillier, R. Bahn and M. Pacold for technical advice on biochemistry and CETSA, S. Parker for GC–MS operation and analysis, D. Jones, R. Rose, Y. Siu and A. Korman for UTP-targeted LC–MS analysis, M. Becerra Flores for ICM-Pro operation, P. Lopez and F. Joseph for FACS analysis, M. Pagano and V. Donato for advice on cell cycle analysis, P. A. Bullock, L. Wu, I. Mohr, C. Y. Park and members of the R.P. laboratory for general discussion, and V. Rubio for discussion on CPS structure, enzymology and allostery. plentiCRISPR v2 was a gift from F. Zhang. Funding was provided by National Institutes of Health grants T32CA009161 (supporting J.S.) R01CA214948 (to R.P.) and R01GM132491 (to R.P.), Pew Charitable Trusts (to R.P.), Alexander and Margaret Stewart Trust (to R.P.) and the American Cancer Society (to R.P.). The metabolomics laboratory was supported by National Institute of Health grant P30CA016087.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R.P. and J.S. Data curation, J.S. Funding acquisition, R.P. Investigation, J.S. Cell cycle analysis, J.S. Bioinformatics, J.S. and H.M. CAD modelling, J.S. and T.J.C. CAD oligomerization, J.S. and B.D.D. CETSA, J.S. and K.M.F. Methodology, R.P. and J.S. Supervision, R.P. Writing—original draft, R.P. and J.S. Writing—review and editing, R.P., J.S. and T.J.C.

Corresponding author

Correspondence to Richard Possemato.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Santiago Ramon Maiques and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Alfredo Giménez-Cassina, in collaboration with the Nature Metabolism team

Additional information

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

Extended data

Extended Data Fig. 1 Acquisition of cell populations at different phases of the cell cycle for targeted metabolomics or for measurement of isotopically labeled pyrimidine intermediates.

a,b, Schematic diagrams of DTB (a), or NOC block (b) cell synchronization procedures employed. c, Schematic diagram showing experimental overview of cell cycle metabolomics. d,e, Metabolic pathways showing participation of extracellular glucose and glutamine into de novo pyrimidine biosynthesis, highlighted (red) shows exogenously supplemented amide-15N-glutamine. f,g, Estimation of relative concentrations (top) or fraction enrichments (bottom) of amide-15N-glutamine-derived metabolites shown in (e) upon DTB release (f), or NOC release (g). h, Measurement of thymine peak height by GC-MS. i, Diagram from (d) with highlighted (red) pathway, exogenously supplemented α-15N-glutamine. j, Estimation of relative concentrations (top) or fraction enrichments (bottom) of α-15N-glutamine-derived metabolites shown in (i) upon the time course sampling of cells post-DTB release. 1 h isotopic labeling. k, Diagram from (d) with highlighted (red) pathway, exogenously supplemented uniformly labeled-13C-glucose. 3 h isotopic labeling. l, Estimation of relative concentrations (top) or fraction enrichments (bottom) of uniformly labeled-13C-glucose-derived metabolites shown in (k) upon the time course sampling of cells post-DTB release. m, A 2-dimensional plot of cells distributed by intracellular complexity (granularity; the Side Scatter (SSC) parameter measured by flow cytometer) and DAPI-stained genomic DNA. 7 different gates constructed for acquisition of cells at different phases of the cell cycle as shown in (n). o, An experimental diagram showing a cell sorting strategy for acquisition of cells at different phases of the cell cycle, primarily targeting cells through the 4 gates (#1, 3, 4 and 7). This strategy includes asynchronous cells supplemented with amide-15N-glutamine (1 h) prior to a fluorescence-activated cell sorting (FACS; Hoechst 33342, 10 µM, 20 h). p, Estimation of relative concentrations of amide-15N-glutamine-derived de novo pyrimidine biosynthetic intermediates. Data are the mean ± s.d. (n = 3, biological replicates). Two-tailed Student’s t-test. *P < 0.1, **P < 0.01, ***P < 0.001, ****P < 0.0001, or NS.

Extended Data Fig. 2 Verification of cell cycle synchronization of wild-type HeLa cells.

a, western blots of canonical cell cycle indicating proteins, cyclin F and phosphorylated histone H3, and de novo pyrimidine proteins of interest, CAD and DHODH. b, Measurements of cell parameters after DTB release: cell number, mean cell volume, and total cell/culture volume in absolute or relative values. Data are the mean ± s.d. (n = 3, technical replicates, 5 independent experiments). c, EdU incorporation of cells synchronized by DTB. d, Volumetric distribution of cells synchronized by DTB.

Source data

Extended Data Fig. 3 Differential accumulations of lipid species post-DTB release.

a,b, Positive ion (a), or negative ion (b) lipidomics of wild-type HeLa cells synchronized by DTB. c, 3–6 h post-DTB-specific accumulations of lipid species. Data are the mean ± s.d. (n = 3, biological replicates). d, Differential accumulation of di- (left) or triglycerides (right) of wild-type HeLa cells synchronized by DTB.

Extended Data Fig. 4 Cell cycle-independent transcription of CAD and CAD co-essential pyrimidine metabolic genes.

a, Top 100 CAD co-dependent genes (DepMap, Broad Institute). b, CRISPR-mediated knock-out effect of CAD co-essential pyrimidine genes on numerous cancer cell lines (DepMap, Broad Institute). UCK1, a cell non-essential gene, and RRM2, a cell-essential gene, are also laid out as extreme examples. c, A curated human pyrimidine metabolism map (modified KEGG). Allosteric activation by PRPP is noted in a green dotted-arrow whereas allosteric inhibition by UTP is in a red dotted-pole. d, Relative transcription levels of CAD co-dependent pyrimidine genes (Cyclebase 3.0). RRM2 and CCNF (cyclin F) are examples of genes known with cell cycle-dependent transcription level change.

Extended Data Fig. 5 Prediction of glycerol gradient-sedimented CAD oligomers, and observation of cell cycle-independent CAD oligomerization.

a,b, Coomassie blue- (a) or silver-stained (b) 4–12% Bis-Tris gel, loaded with a glycerol gradient molecular standard mixture of thyroglobulin, apoferritin, β-amylase, and bovine serum albumin differentially resolved through glycerol gradient rate zonal centrifugation. The results were reproducible over 5 times ((a) and (b) are independent). c, Relative abundance of the silver-stained standards in (b). d, Estimation of glycerol gradient fractions corresponding to CAD oligomers by interpolating CAD oligomer molecular sizes into a regression line generated by the sedimentation information of standard molecules. e, A crystal structure of aspartate transcarbamoylase (ATCase) domain of CAD with R2187 residue highlighted in red (PDB: 5G1O; apo). Each monomer of the ATCase trimer is distinctly colored. f, A crystal structure of dihydroorotase (DHOase) domain of CAD with M1601 residue highlighted in red and pink (PDB: 4C6D; N-carbamoyl-L-aspartate (substrate) bound). Each monomer of the DHOase dimer is distinctly colored. g, Western blots of CAD differentially expressed in R2187A, M1601E, or R2187A/M1601E CAD mutant-expressing sgCAD single cell clones selected. The single cell clones with a red asterisk were mainly tested for CAD activity estimation in this report. h, Western blots of CAD of DTB-synchronized wild-type HeLa cell lysates differentially sedimented throughout a 10–35 % glycerol gradient by rate-zonal centrifugation (left). Quantification of CAD protein intensity from the western blotting results (right). i, Western blots of CAD of NOC-synchronized wild-type HeLa cell lysates differentially sedimented throughout a 10–35 % glycerol gradient by rate-zonal centrifugation (left). Quantification of CAD protein intensity from the western blotting results (right).

Source data

Extended Data Fig. 6 Verification of cell cycle synchronization of G-Loop1 CAD mutant cells and estimation of their CAD activity during the cell cycle.

a,b, Cell parameter measurement (a) and EdU incorporation (b) of wild-type HeLa cells synchronized by NOC. c,d, Cell parameter measurement (c) and (d) EdU incorporation (d) of wild-type HeLa cells synchronized by DTB. e, Estimation of CAD activity of wild-type HeLa cells synchronized by DTB. f,g, Cell parameter measurement (f) and EdU incorporation (g) of G-Loop1 HeLa cells synchronized by NOC. h,i, Cell parameter measurement (h) and EdU incorporation (i) of G-Loop1 HeLa cells synchronized by DTB. j, Estimation of CAD activity of G-Loop1 HeLa cells synchronized by DTB. Data are the mean ± s.d. (n = 3, biological replicates). Two-tailed Student’s t-test. *P < 0.1, **P < 0.01, ***P < 0.001, ****P< 0.0001, or NS.

Extended Data Fig. 7 Verification of identical thermal shifts of wild-type and G-Loop1 CAD upon addition of ATP or CTP.

a, Cellular thermal shift assay (CETSA) of wild-type HeLa in the presence of different amounts of UTP (left), and relative CAD intensities are estimated (right). b, Cellular thermal shift assay (CETSA) of wild-type HeLa, wild-type CAD-reconstituted and G-Loop1-expressing sgCAD cells (left), and relative CAD intensities are estimated (right). No nucleotides were incubated with the cell lysates. c,d, Cellular thermal shift assay (CETSA) of wild-type HeLa or G-Loop1-expressing sgCAD cells in the presence of 30 mM of ATP (c; left) or CTP (d; left), and relative CAD intensities are estimated (c,d; right).

Source data

Extended Data Fig. 8 Verification of CAD mutant protein expression level in selected single cell clone cell lines, and estimation of their CAD activities.

a, Immunoblots of sgCAD single cell clone #1 and 8. Clones with red asterisk used in this report. b, Immunoblots of CAD in wild-type or mutant CAD-expressing sgCAD single cell clones. Wild-type HeLa cell lysate was loaded in titration in western blot for selection of clones with comparable CAD expression. Clones with red asterisk used in this report. c, e, Immunoblots (c) and relative activity (e) of CAD in single cell clones differentially expressing CAD mutants whose mutations correspond to UMP-interacting residues in E. Coli carB. d,f, Immunoblots (d) and relative activity (f) of CAD in single cell clones differentially expressing CAD mutants whose allosteric domain is partially or entirely truncated or glycine-substituted. g,h, Immunoblots (g) and relative activity (h) of CAD in single cell clones differentially expressing CAD mutants whose Loop1 is partially replaced with alanine(s). k, Relative activity of CAD in single cell clones expressing CAD mutants whose three glutamates are substituted for alanines (3E-3A), ± uridine supplementation (100 µg ml-1, 3 h). i,j, Immunoblots (i) and relative activity (j) of CAD in single cell clones differentially expressing CAD mutants whose Loop2 is partially replaced with alanine(s). l,m, Immunoblots (l) and relative activity (m) of CAD in single cell clones differentially expressing CAD mutants whose Loop1 or 2 is duplicated (only glutamates/arginines were mutated; Loop1 3E-3R for Loop1 duplication; Loop2 3R-3E for Loop2 duplication (Loop2 3R-3E immunoblots not shown)) or switched (3E@3R). n, Relative 15N-dihydroorotate concentration of CAD in single cell clones whose three glutamates of Loop1 are replaced with arginines (Loop1 3E-3R). o, Immunoblots of CAD in single cell clones differentially expressing CAD mutants whose T′-loop is replaced with CPS 1T′-loop. Clones with red asterisk used in this report. Data are the mean ± s.d. (n = 3, biological replicates). Two-tailed Student’s t-test vs. wild-type HeLa. *P < 0.1, **P < 0.01, ***P < 0.001, ****P < 0.0001, or NS.

Source data

Extended Data Fig. 9 Proposed mechanisms of UTP-mediated allosteric regulation in wild-type HeLa, G-Loop1, and T′-loop chimera CAD cells and verification of cell cycle synchronization of T′-loop chimera CAD mutant cell line.

a, Structural comparisons of human CAD allosteric domain structure(s) homology-modeled against E. coli carB or human CPS1. Crystal structure of E. coli carB allosteric domain (top left), two human CPS1 allosteric domains (top right; allosteric activator-bound (active state) and apo (inactive state)). Predicted human CAD allosteric domain structures homology-modeled against E. coli carB (bottom left), human CPS1 (bottom right). T′-loop colored green (active), red (inactive). Allosteric ligands shown. Annotated residues exemplify differences in structure between E. coli carB- and human CPS1-based models. b, CAD phylogenetic tree. Uniprot codes in parenthesis. Human CAD, CPS1, and E. coli carB, colored red. c, Overview of pairwise alignment. Percentage identity noted below. d, Docking simulations of UTP-relevant metabolites in human CPS1-based allosteric domain structure of human CAD. e, Alignment of human CAD CPS2 and CPS1 T′-loops. f, Relative frequencies of residue interactions between T′-loop and other domains of human CAD CPS2. Analyses based upon two structures of human CAD CPS2, predicted by homology modeling against human CPS1 structures (PDB: 5DOU, active; 5DOT, inactive). T′-loop-interacting residues in active state (top) or inactive state (bottom) model quantitated via counting neighboring residues of conserved or divergent residues of T′-loop. Human CAD T′-loop residues conserved in human CPS1 are green; divergent, red. Allosteric domain of human CAD, grey. g, Proposed mechanism of CAD allosteric regulation. When [UTP] < Kd of UTP for CAD allosteric domain, Loop1 and Loop2 interact with and stabilize T′-loop, leading to substrate channel formation; active CAD. When [UTP] > Kd, UTP binds to allosteric domain and recruits Loop1 and Loop2, destabilizing T′-loop and substrate channel; inactive CAD. In G-Loop1 mutant, Loop1 constantly interacts with T′-loop, inducing T′-loop stability and substrate channel formation; constitutively active CAD. In T′-loop chimera, [UTP] > Kd, UTP binds to the allosteric domain and recruits Loop1 and Loop2. Because of T′-Loop replacement, loop1-T’-loop interactions and stability regulation are perturbed; no UTP-mediated allosteric regulation. h,i, Cell parameter (h) and EdU incorporation (i) of T′-loop chimera, NOC synchronized. Data are the mean ± s.d. (n = 3, technical replicates, 2 independent experiments).

Extended Data Fig. 10 Measurements of uridine auxotrophic proliferation and uridine-dependent CAD activity change of HeLa, RPE-1, A549, and MDA-MB-231 cell lines established upon transduction of indicated CAD mutants.

ac, HeLa. df, RPE-1. gi, A549. jl, MDA-MB-231. a,d,g,j, Immunoblots of CAD in the indicated cell lines expressing indicated CAD mutants. b,e,h,k, Proliferation of indicated CAD-mutant expressing cells in standard conditions, relative to uridine supplemented (100 µg ml-1). Data are the mean ± s.d. (n = 3, technical replicates, 2 independent experiments). Two-tailed Student’s t-test statistical analyses applied. Compared to wild-type counterpart. c,f,i,l, Estimation of 15N-dihydroorotate concentration of indicated CAD mutant cells in the absence and presence of uridine (100 µg ml-1, 1 h). 1 h tracing. Data are the mean ± s.d. (n = 3, biological replicates). Two-tailed Student’s t-test statistical analyses applied. *P < 0.1, **P < 0.01, ***P < 0.001, ****P < 0.0001, or NS.

Source data

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blots of Fig. 1h.

Source Data Fig. 2

Unprocessed western blots of Fig. 2.

Source Data Fig. 4

Unprocessed western blots of Fig. 4.

Source Data Extended Data Fig. 2

Unprocessed western blots of Extended Fig. 2a.

Source Data Extended Data Fig. 5

Unprocessed stained gels of Extended Fig. 5a,b,g.

Source Data Extended Data Fig. 7

Unprocessed western blots of Extended Fig. 7.

Source Data Extended Data Fig. 8

Unprocessed western blots of Extended Fig. 8.

Source Data Extended Data Fig. 10

Unprocessed western blots of Extended Fig. 10.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shin, J., Mir, H., Khurram, M.A. et al. Allosteric regulation of CAD modulates de novo pyrimidine synthesis during the cell cycle. Nat Metab 5, 277–293 (2023). https://doi.org/10.1038/s42255-023-00735-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-023-00735-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing