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:

Structural basis of lipid head group entry to the Kennedy pathway by FLVCR1

Abstract

Phosphatidylcholine and phosphatidylethanolamine, the two most abundant phospholipids in mammalian cells, are synthesized de novo by the Kennedy pathway from choline and ethanolamine, respectively1,2,3,4,5,6. Despite the essential roles of these lipids, the mechanisms that enable the cellular uptake of choline and ethanolamine remain unknown. Here we show that the protein encoded by FLVCR1, whose mutation leads to the neurodegenerative syndrome posterior column ataxia and retinitis pigmentosa7,8,9, transports extracellular choline and ethanolamine into cells for phosphorylation by downstream kinases to initiate the Kennedy pathway. Structures of FLVCR1 in the presence of choline and ethanolamine reveal that both metabolites bind to a common binding site comprising aromatic and polar residues. Despite binding to a common site, FLVCR1 interacts in different ways with the larger quaternary amine of choline in and with the primary amine of ethanolamine. Structure-guided mutagenesis identified residues that are crucial for the transport of ethanolamine, but dispensable for choline transport, enabling functional separation of the entry points into the two branches of the Kennedy pathway. Altogether, these studies reveal how FLVCR1 is a high-affinity metabolite transporter that serves as the common origin for phospholipid biosynthesis by two branches of the Kennedy pathway.

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: Structure of human FLVCR1 in a choline-bound state.
Fig. 2: FLVCR1 bound to endogenous substrates.
Fig. 3: Ethanolamine and choline are transported by FLVCR1 through a common pathway.
Fig. 4: FLVCR1 fuels both branches of the Kennedy pathway.

Similar content being viewed by others

Data availability

Cryo-EM maps have been deposited in the Electron Microscopy Data Bank (EMDB) under the accession codes EMD-42107 (choline-bound FLVCR1), EMD-42108 (ethanolamine-bound FLVCR1), EMD-42109 (endogenous choline-bound FLVCR1), EMD-42110 (endogenous choline-bound FLVCR1, from images collected in 1 mM ethanolamine) and EMD-42111 (endogenous ligand-bound FLVCR1). Atomic coordinates have been deposited in the Protein Data Bank (PDB) under the accession codes 8UBW (choline-bound FLVCR1), 8UBX (ethanolamine-bound FLVCR1), 8UBY (endogenous choline-bound FLVCR1), 8UBZ (endogenous choline-bound FLVCR1, from images collected in 1 mM ethanolamine) and 8UC0 (endogenous ligand-bound FLVCR1). The atomic coordinates of previously published structures of E.coli SotB (PDB 6KKL) in an inward-facing state and E. coli DgoT (PDB 6E9N) in an inward-facing state were used in this study. Co-dependencies between FLVCR1 and all genes computed from CRISPR DepMap Chronos 2023Q2 used in this study were downloaded from https://depmap.org/portal/all. Source data are provided with this paper.

Code availability

The code written to perform the FLVCR1 DepMap coessentiality analysis is available at https://github.com/artemkhan/Coessentiality_DepMAP_FLVCR1.git.

References

  1. Vance, J. E. Phospholipid synthesis and transport in mammalian cells. Traffic 16, 1–18 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Patel, D. & Witt, S. N. Ethanolamine and phosphatidylethanolamine: partners in health and disease. Oxid. Med. Cell. Longev. 2017, 4829180 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Kent, C. Phospholipid metabolism in mammals. Encycl. Biol. Chem. 3, 314–320 (2004).

    Article  CAS  Google Scholar 

  4. Kennedy, E. P. Sailing to Byzantium. Annu. Rev. Biochem. 61, 1–28 (1992).

    Article  CAS  PubMed  Google Scholar 

  5. Gibellini, F. & Smith, T. K. The Kennedy pathway–de novo synthesis of phosphatidylethanolamine and phosphatidylcholine. IUBMB Life 62, 414–428 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Kennedy, E. P. Synthesis of phosphatides in isolated mitochondria: II. Incorporation of choline into lecithin. J. Biol. Chem. 209, 525–535 (1954).

    Article  CAS  PubMed  Google Scholar 

  7. Yanatori, I., Yasui, Y., Miura, K. & Kishi, F. Mutations of FLVCR1 in posterior column ataxia and retinitis pigmentosa result in the loss of heme export activity. Blood Cells Mol. Dis. 49, 60–66 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Rajadhyaksha, A. M. et al. Mutations in FLVCR1 cause posterior column ataxia and retinitis pigmentosa. Am. J. Hum. Genet. 87, 643–654 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Khan, A. A. & Quigley, J. G. Heme and FLVCR-related transporter families SLC48 and SLC49. Mol. Aspects Med. 34, 669–682 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Corbin, K. D. & Zeisel, S. H. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr. Opin. Gastroenterol. 28, 159–165 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kenny, T. C. et al. Integrative genetic analysis identifies FLVCR1 as a plasma-membrane choline transporter in mammals. Cell Metab. https://doi.org/10.1016/j.cmet.2023.04.003 (2023).

  12. Tsuchiya, M., Tachibana, N., Nagao, K., Tamura, T. & Hamachi, I. Organelle-selective click labeling coupled with flow cytometry allows pooled CRISPR screening of genes involved in phosphatidylcholine metabolism. Cell Metab. 35, 1072–1083 (2023).

    Article  CAS  PubMed  Google Scholar 

  13. Kvarnung, M. et al. Mutations in FLVCR2 associated with Fowler syndrome and survival beyond infancy. Clin. Genet. 89, 99–103 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Meyer, E. et al. Mutations in FLVCR2 are associated with proliferative vasculopathy and hydranencephaly-hydrocephaly syndrome (Fowler syndrome). Am. J. Hum. Genet. 86, 471–478 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lalonde, E. et al. Unexpected allelic heterogeneity and spectrum of mutations in Fowler syndrome revealed by next-generation exome sequencing. Hum. Mutat. 31, 918–923 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Drew, D., North, R. A., Nagarathinam, K. & Tanabe, M. Structures and general transport mechanisms by the major facilitator superfamily (MFS). Chem. Rev. 121, 5289–5335 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sauve, S., Williamson, J., Polasa, A. & Moradi, M. Ins and outs of rocker switch mechanism in major facilitator superfamily of transporters. Membranes 13, 462 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yan, N. Structural biology of the major facilitator superfamily transporters. Annu. Rev. Biophys. 44, 257–283 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Zhang, X. C., Zhao, Y., Heng, J. & Jiang, D. Energy coupling mechanisms of MFS transporters. Protein Sci. 24, 1560–1579 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Okuda, T. et al. Identification and characterization of the high-affinity choline transporter. Nat. Neurosci. 3, 120–125 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Ferguson, S. M. et al. Lethal impairment of cholinergic neurotransmission in hemicholinium-3-sensitive choline transporter knockout mice. Proc. Natl Acad. Sci. USA 101, 8762–8767 (2004).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Iwamoto, H., Blakely, R. D. & De Felice, L. J. Na+, Cl, and pH dependence of the human choline transporter (hCHT) in Xenopus oocytes: the proton inactivation hypothesis of hCHT in synaptic vesicles. J. Neurosci. 26, 9851–9859 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mödinger, Y., Schön, C., Wilhelm, M. & Hals, P.-A. Plasma kinetics of choline and choline metabolites after a single dose of SuperbaBoostTM krill oil or choline bitartrate in healthy volunteers. Nutrients 11, 2548 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Garguilo, M. G. & Michael, A. C. Amperometric microsensors for monitoring choline in the extracellular fluid of brain. J. Neurosci. Methods 70, 73–82 (1996).

    Article  CAS  PubMed  Google Scholar 

  25. Brehm, R., Lindmar, R. & Löffelholz, K. Muscarinic mobilization of choline in rat brain in vivo as shown by the cerebral arterio-venous difference of choline. J. Neurochem. 48, 1480–1485 (1987).

    Article  CAS  PubMed  Google Scholar 

  26. Bianchi, L. et al. Extracellular levels of amino acids and choline in human high grade gliomas: an intraoperative microdialysis study. Neurochem. Res. 29, 325–334 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Plagemann, P. G. Choline metabolism and membrane formation in rat hepatoma cells grown in suspension culture. 3. Choline transport and uptake by simple diffusion and lack of direct exchange with phosphatidylcholine. J. Lipid Res. 12, 715–724 (1971).

    Article  CAS  PubMed  Google Scholar 

  28. Oswald, C. et al. Crystal structures of the choline/acetylcholine substrate-binding protein ChoX from Sinorhizobium meliloti in the liganded and unliganded-closed states. J. Biol. Chem. 283, 32848–32859 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Bärland, N. et al. Mechanistic basis of choline import involved in teichoic acids and lipopolysaccharide modification. Sci. Adv. 8, eabm1122 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Holm, L. Dali server: structural unification of protein families. Nucleic Acids Res. 50, W210–W215 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xiao, Q. et al. Visualizing the nonlinear changes of a drug-proton antiporter from inward-open to occluded state. Biochem. Biophys. Res. Commun. 534, 272–278 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Leano, J. B. et al. Structures suggest a mechanism for energy coupling by a family of organic anion transporters. PLoS Biol. 17, e3000260 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Quigley, J. G. et al. Identification of a human heme exporter that is essential for erythropoiesis. Cell 118, 757–766 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jackson, B. T. Identification of metabolic networks by genetic co-essentiality analysis. Nat. Rev. Mol. Cell Biol. 24, 378 (2023).

    Article  CAS  PubMed  Google Scholar 

  37. Lykidis, A., Wang, J., Karim, M. A. & Jackowski, S. Overexpression of a mammalian ethanolamine-specific kinase accelerates the CDP-ethanolamine pathway. J. Biol. Chem. 276, 2174–2179 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Vermeulen, P. S., Geelen, M. J. H. & van Golde, L. M. G. Substrate specificity of CTP:phosphoethanolamine cytidylyltransferase purified from rat liver. Biochim. Biophys. Acta Lipids Lipid Metab. 1211, 343–349 (1994).

    Article  CAS  Google Scholar 

  39. Taylor, A., Grapentine, S., Ichhpuniani, J. & Bakovic, M. Choline transporter-like proteins 1 and 2 are newly identified plasma membrane and mitochondrial ethanolamine transporters. J. Biol. Chem. 296, 100604 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Navale, A. M. & Paranjape, A. N. Glucose transporters: physiological and pathological roles. Biophys. Rev. 8, 5–9 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Goehring, A. et al. Screening and large-scale expression of membrane proteins in mammalian cells for structural studies. Nat. Protoc. 9, 2574–2585 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  43. Suloway, C. et al. Fully automated, sequential tilt-series acquisition with Leginon. J. Struct. Biol. 167, 11–18 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

  45. Punjani, A., Zhang, H. & Fleet, D. J. Non-uniform refinement: adaptive regularization improves single-particle cryo-EM reconstruction. Nat. Methods 17, 1214–1221 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Scheres, S. H. W. in The Resolution Revolution: Recent Advances in cryoEM Vol. 579 (ed. Crowther, R. A.) 125–157 (Academic, 2016).

  47. Terwilliger, T. C., Ludtke, S. J., Read, R. J., Adams, P. D. & Afonine, P. V. Improvement of cryo-EM maps by density modification. Nat. Methods 17, 923–927 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Jamali, K. et al. Automated model building and protein identification in cryo-EM maps. Nature 628, 450–457 (2024).

  49. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Pavelka, A. et al. CAVER: algorithms for analyzing dynamics of tunnels in macromolecules. IEEE/ACM Trans. Comput. Biol. Bioinform. 13, 505–517 (2016).

    Article  PubMed  Google Scholar 

  54. 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 

  55. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. J. de la Cruz and the Simons Electron Microscopy Center staff for help with data acquisition; the Memorial Sloan Kettering Cancer Center High Performance Computing group for assistance with data processing; the members of the laboratories for comments on the manuscript; and L. Finley for discussions. R.K.H. is supported by the National Institutes of Health (NIH) National Cancer Institute Cancer Center Support Grant P30-CA008748 and is a Searle Scholar. T.C.K. is supported by the NIH National Institute of Diabetes and Digestive and Kidney Diseases (F32 DK127836), the Shapiro-Silverberg Fund for the Advancement of Translational Research and a Merck Postdoctoral Fellowship at The Rockefeller University. K.B. is supported by the NIH National Institute of Diabetes and Digestive and Kidney Diseases (R01 DK123323-01), and a Mark Foundation Emerging Leader Award, and is a Searle Scholar and a Pew-Stewart Scholar. Some of this work was carried out at the Simons Electron Microscopy Center at the New York Structural Biology Center, with major support from the Simons Foundation (SF349247).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Y.S. and R.K.H.; methodology: Y.S., T.C.K., A.K., K.B. and R.K.H.; formal analysis: Y.S. and R.K.H.; investigation, Y.S., T.C.K., K.B. and R.K.H.; writing (original draft): Y.S. and R.K.H.; funding acquisition, T.C.K., K.B. and R.K.H.

Corresponding author

Correspondence to Richard K. Hite.

Ethics declarations

Competing interests

R.K.H. is a consultant for F. Hoffmann-La Roche. K.B. is a scientific adviser to Nanocare Pharmaceuticals and Atavistik Bio. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Camilo Perez and Terry Smith 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 Regulation of FLVCR1-mediated choline uptake.

a, Background subtracted uptake of 20 nM [methyl-3H]choline by vesicles containing FLVCR1 after 30 min in buffer at pH 4.5-10.5. Background was measured by uptake of 20 nM [methyl-3H]choline into the protein-free control liposomes. n = 3 technically independent samples. b, Background subtracted uptake of 20 nM [methyl-3H]choline by FLVCR1 proteoliposome after 30 min in buffer in the presence or absence of Na+, K+, Ca2+, or Mg2+. Background was measured by uptake of 20 nM [methyl-3H]choline into the protein-free control liposomes. n = 3 technically independent samples.

Source Data

Extended Data Fig. 2 Validation of cryo-EM structures of FLVCR1.

a-c Representative cryo-EM images and two-dimensional class averages of human FLVCR1 in 1 mM choline (a), 1 mM ethanolamine (b) or without added substrate (c). Number of collected cryo-EM images are shown. d-h, Plots showing Fourier shell correlations between two independent half-maps (black) and between density-modified map and refined atomic model (red) for choline-bound FLVCR1 (d), ethanolamine-bound FLVCR1 (e), endogenous choline-bound FLVCR1 obtained from FLVCR1 incubated with 1 mM ethanolamine (f), endogenous choline-bound FLVCR1 obtained from FLVCR1 without exogenous ligand incubation (g), and endogenous ligand-bound FLVCR1 obtained from FLVCR1 without exogenous ligand incubation (h). Dashed lines are indicated at FSC = 0.5 and FSC = 0.143. i-m, Local resolution plots of choline-bound FLVCR1 (i), ethanolamine-bound FLVCR1 (j), endogenous-choline bound FLVCR1 obtained from FLVCR1 incubated with 1 mM ethanolamine (k), endogenous choline-bound FLVCR1 obtained from FLVCR1 without exogenous substrate incubation (l), and endogenous ligand-bound FLVCR1 obtained from FLVCR1 without exogenous ligand incubation (m).

Source Data

Extended Data Fig. 3 Cryo-EM analysis of hFLVCR1.

Flow-chart summarizing Cryo-EM image acquisition and processing of human FLVCR1 without added substrate (a), in 1 mM choline (b), or 1 mM ethanolamine (c). Number of collected cryo-EM images and selected particles are shown.

Extended Data Fig. 4 Choline-bound FLVCR1 adopts an inward-facing conformation.

a, Superposition of TM1-6 and TM7-12. RMSD = 3.5 Å. b, Central cavity viewed from the cytosolic side. Aspartate and glutamate residues in and near the entrance to the central cavity are shown as sticks. c, Central section of choline-bound FLVCR1 is contiguous with choline shown as sticks. d, Blue spheres depict the minimum radius of the central cavity in the choline-bound state as a function of position. e-f, Superposition of choline-bound FLVCR1 (blue) with E. coli SotB (D; PDB: 6KKL; RMSD = 2.3 Å; red) (e) or E. coli DgoT (E; PDB: 6E9N; RMSD = 2.6 Å; cyan) (f).

Extended Data Fig. 5 Sequence alignment of SLC49A family members and identified disease-associated substitutions.

a, Sequence alignment of human FLVCR1 (SLC49A1), human FLVCR2 (SLC49A2), human MFSD7 (SLC49A3), human DIRC2 (SLC49A4) and Drosophila CG1358 performed using Clustal Omega54 and pyBoxshade (https://github.com/mdbaron42/pyBoxshade). Substrate-binding site residues are highlighted by red boxes. Residues whose substitution leads to PCARP or Fowler syndrome are highlighted by blue and green boxes, respectively. b, Structure of choline-bound FLVCR1 (left) and Alphafold2 model of FLVCR2 (right)55 with residues whose mutation leads to PCARP and Fowler syndrome highlighted in blue and green, respectively. FLVCR1 substrate binding site residues and the corresponding residues in FLVCR2 are highlighted in red.

Extended Data Fig. 6 Cryo-EM structures of FLVCR1 determined in a substrate-free condition.

a-b, Cryo-EM density maps and atomic models of FLVCR1 in endogenous choline (a) and endogenous ligand-bound (b) states. c-d, Central slice of the central cavity of choline-bound (c) and endogenous ligand-bound (d) states, with surface colored white. Choline is shown as sticks in c. Endogenous ligand is not modeled or shown in d. e, Superposition of choline-bound (grey) and endogenous choline-bound (blue) states. f, Superposition of substrate-binding sites in choline-bound (grey) and endogenous choline-bound (blue) states.

Extended Data Fig. 7 Cryo-EM structures of FLVCR1 determined in 1mM ethanolamine.

a-b, Cryo-EM density maps of endogenous choline-bound FLVCR1 (a) and ethanolamine-bound FLVCR1 (b) states, determined from particles imaged in the presence of 1 mM ethanolamine. c, Superposition of ethanolamine-bound (gold) and choline-bound (blue) states. d, Substrate-binding site in endogenous choline-bound FLVCR1 state, determined from particles imaged in the presence of 1 mM ethanolamine. Residues and modelled substrates are shown as sticks. Density is shown as a grey isosurface and contoured at 3.0 σ. e-f, Coordination of ethanolamine in the substrate-binding site in ethanolamine-bound state (e) and choline in the substrate-binding site in the choline-bound state (f). Polar interactions are shown as dashed lines and distances as solid lines.

Extended Data Fig. 8 FLVCR1 substrate-binding site mutants.

a, FSEC analysis of wild-type or substrate-binding site mutants fused to mCerulean expressed in FLVCR1-knockout HEK293T cells. b, Western blot analysis of FLVCR1-knockout HEK293T cells expressing a vector control or wild-type or mutant FLVCR1 cDNA. GAPDH was ran on a separate gel as sample processing controls. For western blot source data, see Supplementary Fig. 1. c, Cumulative log2 fold change in cell number of HEK293T cells and FLVCR1-knockout HEK293T cells expressing a vector control or wild-type or mutant FLVCR1 cDNA. n = 3 biologically independent samples. (P-values: HEK293T WT and HEK293T FLVCR1 KO + FLVCR1 WT cDNA; p = 3x10−3, HEK293T FLVCR1 KO + FLVCR1 WT cDNA and HEK293T FLVCR1 KO + FLVCR1 Q214A cDNA; p = 9x10−1, HEK293T FLVCR1 KO + Vector and HEK293T FLVCR1 KO + FLVCR1 W125A cDNA; p = 9x10−1, HEK293T FLVCR1 KO + Vector and HEK293T FLVCR1 KO + FLVCR1 Y153A cDNA; p = 1x10−1, HEK293T FLVCR1 KO + FLVCR1 WT cDNA and HEK293T FLVCR1 KO + Vector; p = 8x10−5).

Source Data

Extended Data Fig. 9 Incorporation of labeled choline into betaine requires FLVCR1.

Schematic for tracing [1,2-13C2] choline into betaine (left) and the abundance of betaine M + 2 after incubation with 21.5 µM [1,2-13C2] choline for 1 h in FLVCR1-knockout HEK293T cells expressing a vector control or wild-type or mutant FLVCR1 cDNA (right). n = 3 biologically independent samples.

Source Data

Extended Data Table 1 Cryo-EM data collection, refinement, and validation statistics

Supplementary information

Supplementary Fig. 1

Raw, uncropped western blot image of Extended Data Fig. 8b. GAPDH was run on a separate gel as a sample processing control.

Reporting Summary

Peer Review File

Source data

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

Son, Y., Kenny, T.C., Khan, A. et al. Structural basis of lipid head group entry to the Kennedy pathway by FLVCR1. Nature 629, 710–716 (2024). https://doi.org/10.1038/s41586-024-07374-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07374-4

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

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