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:

De novo lipogenesis is essential for platelet production in humans

Abstract

Acetyl-CoA carboxylase (ACC) catalyses the first step of de novo lipogenesis (DNL). Pharmacologic inhibition of ACC has been of interest for therapeutic intervention in a wide range of diseases. We demonstrate here that ACC and DNL are essential for platelet production in humans and monkeys, but in not rodents or dogs. During clinical evaluation of a systemically distributed ACC inhibitor, unexpected dose-dependent reductions in platelet count were observed. While platelet count reductions were not observed in rat and dog toxicology studies, subsequent studies in cynomolgus monkeys recapitulated these platelet count reductions with a similar concentration response to that in humans. These studies, along with ex vivo human megakaryocyte maturation studies, demonstrate that platelet lowering is a consequence of DNL inhibition likely to result in impaired megakaryocyte demarcation membrane formation. These observations demonstrate that while DNL is a minor quantitative contributor to global lipid balance in humans, DNL is essential to specific lipid pools of physiological importance.

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: Administration of PF-05175157 reversibly and dose-dependently lowered platelet count in healthy human participants.
Fig. 2: Airyscan confocal imaging characterizing ex vivo cord-blood-derived human MK culture system on day 12 of culture.
Fig. 3: ACC inhibition decreases platelet production in human MKs in vitro.
Fig. 4: DNL is required for late-stage MK maturation and platelet formation.
Fig. 5: Large, surface-complex MKs (high SSC) are decreased from the mature population in ACC inhibitor-treated cells.
Fig. 6: ACC inhibitor treatment blocks DNL and reduces PCs composed of DNL-derived fatty acids.
Fig. 7: Decreased platelets and effect on bone marrow MK after administration of ACC inhibitors to monkeys.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. Expression data for murine and human megakaryocytes were obtained from the European Bioinformatics Institute Gene Expression Atlas. For murine megakaryocytes RNA-seq data from experiment E-MTAB-3079 (ref. 59) were obtained. For human megakaryocytes data from experiment E-MTAB-3827 (ref. 60) were obtained. Source data and the relevant protocols can be found at: https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-3079/ and https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-3827/.

References

  1. Donnelly, K. L. et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Invest. 115, 1343–1351 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Hellerstein, M. K. De novo lipogenesis in humans: metabolic and regulatory aspects. Eur. J. Clin. Nutr. 53 (Suppl. 1), S53–S65 (1999).

    PubMed  Google Scholar 

  3. Strawford, A., Antelo, F., Christiansen, M. & Hellerstein, M. K. Adipose tissue triglyceride turnover, de novo lipogenesis, and cell proliferation in humans measured with 2H2O. Am. J. Physiol. Endocrinol. Metab. 286, E577–E588 (2004).

    CAS  PubMed  Google Scholar 

  4. Timlin, M. T. & Parks, E. J. Temporal pattern of de novo lipogenesis in the postprandial state in healthy men. Am. J. Clin. Nutr. 81, 35–42 (2005).

    CAS  PubMed  Google Scholar 

  5. Esler, W. P. et al. Human sebum requires de novo lipogenesis, which is increased in acne vulgaris and suppressed by acetyl-CoA carboxylase inhibition. Sci. Transl. Med. 11, eaau8465 (2019).

    CAS  PubMed  Google Scholar 

  6. Harwood, H. J. Jr. Treating the metabolic syndrome: acetyl-CoA carboxylase inhibition. Expert Opin. Ther. Targets 9, 267–281 (2005).

    CAS  PubMed  Google Scholar 

  7. Lambert, J. E., Ramos-Roman, M. A., Browning, J. D. & Parks, E. J. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology 146, 726–735 (2014).

    CAS  PubMed  Google Scholar 

  8. Menendez, J. A. & Lupu, R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer 7, 763–777 (2007).

    CAS  PubMed  Google Scholar 

  9. Samuel, V. T. & Shulman, G. I. The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux. J. Clin. Invest. 126, 12–22 (2016).

    PubMed  PubMed Central  Google Scholar 

  10. Saggerson, D. Malonyl-CoA, a key signaling molecule in mammalian cells. Annu. Rev. Nutr. 28, 253–272 (2008).

    CAS  PubMed  Google Scholar 

  11. Waite, M. & Wakil, S. J. Studies on the mechanism of fatty acid synthesis. XII. Acetyl coenzyme A carboxylase. J. Biol. Chem. 237, 2750–2757 (1962).

    CAS  PubMed  Google Scholar 

  12. McGarry, J. D., Mannaerts, G. P. & Foster, D. W. A possible role for malonyl-CoA in the regulation of hepatic fatty acid oxidation and ketogenesis. J. Clin. Invest. 60, 265–270 (1977).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Tong, L. & Harwood, H. J. Jr. Acetyl-coenzyme A carboxylases: versatile targets for drug discovery. J. Cell Biochem. 99, 1476–1488 (2006).

    CAS  PubMed  Google Scholar 

  14. Corbett, J. W. et al. Discovery of small molecule isozyme non-specific inhibitors of mammalian acetyl-CoA carboxylase 1 and 2. Bioorg. Med. Chem. Lett. 20, 2383–2388 (2010).

    CAS  PubMed  Google Scholar 

  15. Freeman-Cook, K. D. et al. Maximizing lipophilic efficiency: the use of Free–Wilson analysis in the design of inhibitors of acetyl-CoA carboxylase. J. Med. Chem. 55, 935–942 (2012).

    CAS  PubMed  Google Scholar 

  16. Griffith, D. A. et al. Spirolactam-based acetyl-CoA carboxylase inhibitors: toward improved metabolic stability of a chromanone lead structure. J. Med. Chem. 56, 7110–7119 (2013).

    CAS  PubMed  Google Scholar 

  17. Griffith, D. A. et al. Decreasing the rate of metabolic ketone reduction in the discovery of a clinical acetyl-CoA carboxylase inhibitor for the treatment of diabetes. J. Med. Chem. 57, 10512–10526 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Harwood, H. J. Jr. et al. Isozyme-nonselective N-substituted bipiperidylcarboxamide acetyl-CoA carboxylase inhibitors reduce tissue malonyl-CoA concentrations, inhibit fatty acid synthesis, and increase fatty acid oxidation in cultured cells and in experimental animals. J. Biol. Chem. 278, 37099–37111 (2003).

    CAS  PubMed  Google Scholar 

  19. Kim, C. W. et al. Acetyl CoA carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metab. 26, 576 (2017).

    CAS  PubMed  Google Scholar 

  20. Stiede, K. et al. Acetyl-coenzyme A carboxylase inhibition reduces de novo lipogenesis in overweight male subjects: a randomized, double-blind, crossover study. Hepatology 66, 324–334 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Becker, R. C., Sexton, T. & Smyth, S. S. Translational implications of platelets as vascular first responders. Circ. Res. 122, 506–522 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Thon, J. N. & Italiano, J. E. Platelet formation. Semin. Hematol. 47, 220–226 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Geddis, A. E. Megakaryopoiesis. Semin. Hematol. 47, 212–219 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Eckly, A. et al. Biogenesis of the demarcation membrane system (DMS) in megakaryocytes. Blood 123, 921–930 (2014).

    CAS  PubMed  Google Scholar 

  25. Italiano, J. E. Jr., Lecine, P., Shivdasani, R. A. & Hartwig, J. H. Blood platelets are assembled principally at the ends of proplatelet processes produced by differentiated megakaryocytes. J. Cell Biol. 147, 1299–1312 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. White, M. J. & Kile, B. T. Apoptotic processes in megakaryocytes and platelets. Semin. Hematol. 47, 227–234 (2010).

    CAS  PubMed  Google Scholar 

  27. Lepropre, S. et al. AMPK-ACC signaling modulates platelet phospholipids and potentiates thrombus formation. Blood 132, 1180–1192 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Debili, N. et al. Different expression of CD41 on human lymphoid and myeloid progenitors from adults and neonates. Blood 97, 2023–2030 (2001).

    CAS  PubMed  Google Scholar 

  29. Poujol, C., Ware, J., Nieswandt, B., Nurden, A. T. & Nurden, P. Absence of GPIbɑ is responsible for aberrant membrane development during megakaryocyte maturation: ultrastructural study using a transgenic model. Exp. Hematol. 30, 352–360 (2002).

    CAS  PubMed  Google Scholar 

  30. Barnes, D., et al. Cyclohexane derivatives as inhibitors of acetyl-CoA carboxylase. Patent WO 2011/067306 (Novartis AG, Switzerland, 2011).

  31. Liu, N., et al. Cyclic ketoenols for therapy. US patent 2012/0129833 (Bayer Pharma Aktiengesellschaft, Germany, 2012).

  32. Loomba, R. et al. GS-0976 reduces hepatic steatosis and fibrosis markers in patients with nonalcoholic fatty liver disease. Gastroenterology 155, 1463–1473.e6 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Lin, V. C. et al. Osthole suppresses fatty acid synthase expression in HER2-overexpressing breast cancer cells through modulating Akt/mTOR pathway. J. Agric. Food Chem. 58, 4786–4793 (2010).

    CAS  PubMed  Google Scholar 

  34. Romero, F. A., Jones, C. T., Xu, Y., Fenaux, M. & Halcomb, R. L. The race to bash NASH: emerging targets and drug development in a complex liver disease. J. Med. Chem. 63, 5031–5073 (2020).

    CAS  PubMed  Google Scholar 

  35. De Botton, S. et al. Platelet formation is the consequence of caspase activation within megakaryocytes. Blood 100, 1310–1317 (2002).

    PubMed  Google Scholar 

  36. Schulze, H. et al. Characterization of the megakaryocyte demarcation membrane system and its role in thrombopoiesis. Blood 107, 3868–3875 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Corash, L., Shafer, B. & Perlow, M. Heterogeneity of human whole blood platelet subpopulations. II. Use of a subhuman primate model to analyze the relationship between density and platelet age. Blood 52, 726–734 (1978).

    CAS  PubMed  Google Scholar 

  38. Bergman, A. et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of a liver-targeting Acetyl-CoA carboxylase inhibitor (PF-05221304): a three-part randomized phase 1 study. Clin. Pharm. Drug Dev. 9, 514–526 (2020).

    CAS  Google Scholar 

  39. Schick, P. K., Williams-Gartner, K. & He, X. L. Lipid composition and metabolism in megakaryocytes at different stages of maturation. J. Lipid Res. 31, 27–35 (1990).

    CAS  PubMed  Google Scholar 

  40. Scott, K. E. et al. Metabolic regulation of invadopodia and invasion by acetyl-CoA carboxylase 1 and de novo lipogenesis. PLoS ONE 7, e29761 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Jensen, K. K. et al. Dose-dependent effects of siRNA-mediated inhibition of SCAP on PCSK9, LDLR, and plasma lipids in mouse and rhesus monkey. J. Lipid Res. 57, 2150–2162 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hellerstein, M. K. et al. Measurement of de novo hepatic lipogenesis in humans using stable isotopes. J. Clin. Invest. 87, 1841–1852 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Arriaga, M., South, K., Cohen, J. L. & Mazur, E. M. Interrelationship between mitosis and endomitosis in cultures of human megakaryocyte progenitor cells. Blood 69, 486–492 (1987).

    CAS  PubMed  Google Scholar 

  44. Mattia, G. et al. Different ploidy levels of megakaryocytes generated from peripheral or cord blood CD34+ cells are correlated with different levels of platelet release. Blood 99, 888–897 (2002).

    CAS  PubMed  Google Scholar 

  45. Mwamtemi, H. H., Higuchi, T., Sawai, N., Hidaka, E. & Koike, K. Quantitative and qualitative differences in thrombopoietin-dependent hematopoietic progenitor development between cord blood and bone marrow. Transplantation 69, 1645–1654 (2000).

    CAS  PubMed  Google Scholar 

  46. Pineault, N., Robert, A., Cortin, V. & Boyer, L. in Basic Cell Culture Protocols. Methods in Molecular Biology (Methods and Protocols) Vol. 946 (eds Helgason, C. & Miller, C.) 205–224 (Humana Press, 2012).

  47. Pluthero, F. G. & Kahr, W. H. A. Imaging platelets and megakaryocytes by high-resolution laser fluorescence microscopy. Methods Mol. Biol. 1812, 13–31 (2018).

    CAS  PubMed  Google Scholar 

  48. Futatsugi, K. et al. Discovery and optimization of imidazopyridine-based inhibitors of diacylglycerol acyltransferase 2 (DGAT2). J. Med. Chem. 58, 7173–7185 (2015).

    CAS  PubMed  Google Scholar 

  49. Damen, C. W., Isaac, G., Langridge, J., Hankemeier, T. & Vreeken, R. J. Enhanced lipid isomer separation in human plasma using reversed-phase UPLC with ion-mobility/high-resolution MS detection. J. Lipid Res. 55, 1772–1783 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Bird, S. S., Marur, V. R., Sniatynski, M. J., Greenberg, H. K. & Kristal, B. S. Lipidomics profiling by high-resolution LC-MS and high-energy collisional dissociation fragmentation: focus on characterization of mitochondrial cardiolipins and monolysocardiolipins. Anal. Chem. 83, 940–949 (2011).

    CAS  PubMed  Google Scholar 

  51. Clasquin, M. F., Melamud, E. & Rabinowitz, J. D. LC-MS data processing with MAVEN: a metabolomic analysis and visualization engine. Curr. Protoc. Bioinformatics 37, 14.11 (2012).

    Google Scholar 

  52. Melamud, E., Vastag, L. & Rabinowitz, J. D. Metabolomic analysis and visualization engine for LC-MS data. Anal. Chem. 82, 9818–9826 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Hellerstein, M. K. & Neese, R. A. Mass isotopomer distribution analysis at eight years: theoretical, analytic, and experimental considerations. Am. J. Physiol. 276, E1146–E1170 (1999).

    CAS  PubMed  Google Scholar 

  54. Turner, S. M. et al. Dissociation between adipose tissue fluxes and lipogenic gene expression in ob/ob mice. Am. J. Physiol. Endocrinol. Metab. 292, E1101–E1109 (2007).

    CAS  PubMed  Google Scholar 

  55. Price, J. C. et al. Measurement of human plasma proteome dynamics with 2H2O and liquid chromatography tandem mass spectrometry. Anal. Biochem. 420, 73–83 (2012).

    CAS  PubMed  Google Scholar 

  56. Shah, V., Herath, K., Previs, S. F., Hubbard, B. K. & Roddy, T. P. Headspace analyses of acetone: a rapid method for measuring the 2H-labeling of water. Anal. Biochem. 404, 235–237 (2010).

    CAS  PubMed  Google Scholar 

  57. Jungas, R. L. Fatty acid synthesis in adipose tissue incubated in tritiated water. Biochemistry 7, 3708–3717 (1968).

    CAS  PubMed  Google Scholar 

  58. Tomer, A. et al. Flow cytometric analysis of megakaryocytes from patients with abnormal platelet counts. Blood 74, 594–601 (1989).

    CAS  PubMed  Google Scholar 

  59. Qian, P. et al. The Dlk1-Gtl2 locus preserves LT-HSC function by inhibiting the PI3K-mTOR pathway to restrict mitochondrial metabolism. Cell Stem Cell 18, 214–228 (2016).

    CAS  PubMed  Google Scholar 

  60. Adams, D. et al. BLUEPRINT to decode the epigenetic signature written in blood. Nat. Biotechnol. 30, 224–226 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Griffith, A. Smith and K. Huard for assistance in sourcing the ACC and FASN inhibitors for testing; E. Pashos for compiling the lipogenic enzyme expression data in rodent and human MKs; and G. El Sebae for help with staining MK cell preparations. We also thank M. Birnbaum for helpful discussions. This work was funded by Pfizer Inc.

Author information

Authors and Affiliations

Authors

Contributions

K.L.K. designed and conducted the ex vivo studies and flow cytometry and interpretation of data. G.E.S., S.C.-G., A.B. and C.B. designed and oversaw clinical studies and analysed clinical data. S.C.-G. also conducted statistical analysis for clinical and ex vivo studies. W.J.R., G.C., T.S. and N.S. designed and oversaw nonhuman primate studies and interpretation of nonhuman primate data. K.L.K., K.H. and S.A. conducted florescence confocal microscopy studies. N.S. contributed to electron microscopy studies. M.C., N.B.V., Z.S. and E.Z.Q. conducted lipidomic analysis. M.C., M.D.M., K.W.L. and M.K.H. contributed to design, conduct, analysis and interpretation of stable isotope studies. P.A.A., T.T.R. and D.B. contributed to ex vivo study design, conduct and interpretation. J.A.P. contributed to study conceptualization and interpretation. W.P.E. conceived, designed and supervised the project; contributed to study design; interpreted data; and wrote the manuscript. K.L.K., W.J.R., G.E.S., M.C., K.W.L. and M.K.H. also contributed to writing the manuscript. All authors contributed to data interpretation and critical review of the manuscript.

Corresponding author

Correspondence to William P. Esler.

Ethics declarations

Competing interests

K.L.K., W.J.R., M.C., K.H., S.A., P.A.A., S.C.-G., N.S., N.B.V., T.T.R., G.C., A.B., C.B., Z.S., E.Z.Q., T.S., J.A.P. and W.P.E. are employees and may be shareholders of Pfizer Inc. G.E.S. and D.B. were employees of Pfizer at the time these studies were conducted and may be shareholders of Pfizer Inc. M.K.H. is a consultant to Pfizer Inc.

Additional information

Peer review information Primary Handling Editors: Elena Bellafante; George Caputa.

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

Extended data

Extended Data Fig. 1 Platelet counts in 4-month oral toxicity studies of PF-05175157 in rats and dogs.

a, platelet counts in male and female Wistar Han rats on day 118. N=10 per group. Wistar Han rats were from Charles River Laboratories 8–9 wks old. b, platelet counts in male and female dogs during pre-study (day -13) and day 114. N=3 per sex per group. Dogs were Beagles (male and female) from Marshal Farms > 8 months old. Data are mean ± standard deviation. Symbols represent individual values.

Extended Data Fig. 2 May-Grünwald Giemsa stained megakaryocytes from ex vivo cultures.

Cytospun slides were prepared from in vitro cultures of megakaryocytes on day 12 of culture. All photos were taken at the same magnification (100x oil) and the scale bars shown on images a and c represent 10 microns. a. Low polyploid megakaryocyte (4N) (arrowhead). b, c, Large cells with multilobulated nuclei and granular cytoplasm are mature megakaryocytes (arrows) (≥16N).

Extended Data Fig. 3 DMS continuity between vehicle and ACCi (10µM) treated groups in lower polyploid MKs.

Megakaryocytes differentiated from bone marrow derived CD34+ cells were utilized to visualize internal DMS continuity between vehicle and ACCi (10µM) treated groups in lower polyploid cells on day 13 of culture (Scale bars=10µm, green= CD41a-FITC/ DMS, Red=α-tubulin, blue=DNA/ DAPI) (left quad panels are 2D images from a single, z-stack optical slice; right enlarged image is a 3D render of the same cell through a similar plane). a, vehicle treated low polyploid MK exhibiting highly contiguous DMS (green) throughout the cytoplasmic volume. b, ACCi treated (10µM) lower polyploid MK showing an apparent reduction with discontinuous and diffuse with less structurally defined DMS staining (green). The 3D merged rendered movies of these cells are available as Supplemental Videos 34.

Extended Data Fig. 4 Expression of lipogenic genes in murine and human megakaryocytes.

Expression data for murine and human megakaryocytes were obtained from the European Bioinformatics Institute (EBI) Gene Expression Atlas. For murine megakaryocytes a, RNAseq data from experiment E-MTAB-307959 from were obtained. Mice were C57BL/6J mice between 4–6 weeks of age. Data represent one biological experiment. For human megakaryocytes b, data from experiment E-MTAB-382760 were obtained. Boxplots of the human megakaryocyte samples (n=2) were produced using the ggplot2 library in R. The lower hinge of the boxplot represents the lower of the two expression values and the higher hinge of the boxplot represent the higher of the two values. Normalized expression values of ACC and FASN were higher in human megakaryocytes compared to their murine counterparts. Source data and the relevant protocols can be found at: https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-3079/ and https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-3827/.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Tables 1–3

Reporting Summary

Supplementary Video 1

Video of vehicle-treated high-ploidy MK cell from Fig. 5c

Supplementary Video 2

Video of ACCi-treated high-ploidy MK cell from Fig. 5d

Supplementary Video 3

Video of vehicle-treated lower-ploidy MK cell from Extended Data Fig. 3a

Supplementary Video 4

Video of ACCi-treated lower-ploidy MK cell from Extended Data Fig. 3b

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kelly, K.L., Reagan, W.J., Sonnenberg, G.E. et al. De novo lipogenesis is essential for platelet production in humans. Nat Metab 2, 1163–1178 (2020). https://doi.org/10.1038/s42255-020-00272-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00272-9

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research