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:

Efficient megakaryopoiesis and platelet production require phospholipid remodeling and PUFA uptake through CD36

Abstract

Lipids contribute to hematopoiesis and membrane properties and dynamics; however, little is known about the role of lipids in megakaryopoiesis. Here we show that megakaryocyte progenitors, megakaryocytes and platelets present a unique lipidome progressively enriched in polyunsaturated fatty acid (PUFA)-containing phospholipids. In vitro, inhibition of both exogenous fatty acid functionalization and uptake as well as de novo lipogenesis impaired megakaryocyte differentiation and proplatelet production. In vivo, mice on a high saturated fatty acid diet had significantly lower platelet counts, which was prevented by eating a PUFA-enriched diet. Fatty acid uptake was largely dependent on CD36, and its deletion in mice resulted in low platelets. Moreover, patients with a CD36 loss-of-function mutation exhibited thrombocytopenia and increased bleeding. Our results suggest that fatty acid uptake and regulation is essential for megakaryocyte maturation and platelet production and that changes in dietary fatty acids may be a viable target to modulate platelet counts.

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: Megakaryocytes and platelets have a unique lipid profile that is enriched in PUFAs.
Fig. 2: Fatty acid incorporation and de novo lipogenesis are necessary for MK differentiation and efficient proplatelet formation.
Fig. 3: A high-saturated-fat diet significantly alters MK phenotype and reduces platelet counts.
Fig. 4: Platelet counts can be modified in vivo by altering dietary PUFA composition.
Fig. 5: Lack of CD36 in mice reduces cellular fatty acid incorporation and impairs proplatelet formation.
Fig. 6: Megakaryocytes and platelet counts in Cd36/ mice are not affected by high-fat diets enriched in fatty acids.
Fig. 7: Identification of a CD36 loss-of-function variant (p.Tyr325Ter) in patients with thrombocytopenia.

Similar content being viewed by others

Data availability

All data supporting the findings in this study are included in the main article and associated files. Source data are provided with this manuscript. All raw lipidomic data can be found at https://www.ebi.ac.uk/metabolights/MTBLS8042.

Code availability

Instructions and code for the automated pipeline analysis of proplatelet production from megakaryocytes can be found at https://github.com/broadinstitute/Italiano-MK-Analysis.

References

  1. van Meer, G., Voelker, D. R. & Feigenson, G. W. Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112–124 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Murphy, A. J. et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J. Clin. Invest. 121, 4138–4149 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bansal, P. et al. Current updates on role of lipids in hematopoiesis. Infect. Disord. Drug Targets 18, 192–198 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Lee, M. K. S., Al-Sharea, A., Dragoljevic, D. & Murphy, A. J. Hand of FATe: lipid metabolism in hematopoietic stem cells. Curr. Opin. Lipidol. 29, 240–245 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Pernes, G., Flynn, M. C., Lancaster, G. I. & Murphy, A. J. Fat for fuel: lipid metabolism in haematopoiesis. Clin. Transl. Immunol. 8, e1098 (2019).

    Article  Google Scholar 

  6. Collins, J. M. et al. De novo lipogenesis in the differentiating human adipocyte can provide all fatty acids necessary for maturation. J. Lipid Res. 52, 1683–1692 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Noetzli, L. J., French, S. L. & Machlus, K. R. New insights into the differentiation of megakaryocytes from hematopoietic progenitors. Arterioscler. Thromb. Vasc. Biol. 39, 1288–1300 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Machlus, K. R. & Italiano, J. E. The incredible journey: from megakaryocyte development to platelet formation. J. Cell Biol. 201, 785–796 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pietras, E. M. et al. Functionally distinct subsets of lineage-biased multipotent progenitors control blood production in normal and regenerative conditions. Cell Stem Cell 17, 35–46 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Whitaker, B., Rajbhandary, S., Kleinman, S., Harris, A. & Kamani, N. Trends in United States blood collection and transfusion: results from the 2013 AABB Blood Collection, Utilization, and Patient Blood Management Survey. Transfusion 56, 2173–2183 (2016).

    Article  PubMed  Google Scholar 

  12. Provan, D. et al. Updated international consensus report on the investigation and management of primary immune thrombocytopenia. Blood Adv. 3, 3780–3817 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Ghanima, W. et al. Bone marrow fibrosis in 66 patients with immune thrombocytopenia treated with thrombopoietin-receptor agonists: a single-center, long-term follow-up. Haematologica 99, 937–944 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Mitchell, W. B. & Bussel, J. B. Thrombopoietin receptor agonists: a critical review. Semin. Hematol. 52, 46–52 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Prica, A., Sholzberg, M. & Buckstein, R. Safety and efficacy of thrombopoietin-receptor agonists in myelodysplastic syndromes: a systematic review and meta-analysis of randomized controlled trials. Br. J. Haematol. 167, 626–638 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Manni, M. M. et al. Acyl chain asymmetry and polyunsaturation of brain phospholipids facilitate membrane vesiculation without leakage. eLife 7, e34394 (2018).

  17. Valet, C. et al. Adipocyte fatty acid transfer supports megakaryocyte maturation. Cell Rep. 32, 107875 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Kelly, K. L. et al. De novo lipogenesis is essential for platelet production in humans. Nat. Metab. 2, 1163–1178 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Machlus, K. R. et al. Synthesis and dephosphorylation of MARCKS in the late stages of megakaryocyte maturation drive proplatelet formation. Blood 127, 1468–1480 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Febbraio, M. et al. A null mutation in murine CD36 reveals an important role in fatty acid and lipoprotein metabolism. J. Biol. Chem. 274, 19055–19062 (1999).

    Article  CAS  PubMed  Google Scholar 

  21. Aitman, T. J. et al. Malaria susceptibility and CD36 mutation. Nature 405, 1015–1016 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Hsieh, F. L. et al. The structural basis for CD36 binding by the malaria parasite. Nat. Commun. 7, 12837 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Escribá, P. V. et al. Membrane lipid therapy: modulation of the cell membrane composition and structure as a molecular base for drug discovery and new disease treatment. Prog. Lipid Res. 59, 38–53 (2015).

    Article  PubMed  Google Scholar 

  24. Doi, O., Doi, F., Schroeder, F., Alberts, A. W. & Vagelos, P. R. Manipulation of fatty acid composition of membrane phospholipid and its effects on cell growth in mouse LM cells. Biochim. Biophys. Acta 509, 239–250 (1978).

    Article  CAS  PubMed  Google Scholar 

  25. Pinot, M. et al. Lipid cell biology. Polyunsaturated phospholipids facilitate membrane deformation and fission by endocytic proteins. Science 345, 693–697 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Hales, C. M., Carroll, M. D., Fryar, C. D. & Ogden, C. L. Prevalence of obesity and severe obesity among adults: United States, 2017–2018. NCHS Data Brief https://www.cdc.gov/nchs/data/databriefs/db360-h.pdf (2020).

  27. Yin, R., Wang, X., Li, K., Yu, K. & Yang, L. Lipidomic profiling reveals distinct differences in plasma lipid composition in overweight or obese adolescent students. BMC Endocr. Disord. 21, 201 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Vilahur, G., Ben-Aicha, S. & Badimon, L. New insights into the role of adipose tissue in thrombosis. Cardiovasc. Res. 113, 1046–1054 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Badimon, L., Hernández Vera, R., Padró, T. & Vilahur, G. Antithrombotic therapy in obesity. Thromb. Haemost. 110, 681–688 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Barale, C. & Russo, I. Influence of cardiometabolic risk factors on platelet function. Int. J. Mol. Sci. 21, 623 (2020).

  31. Haas, S. et al. Inflammation-induced emergency megakaryopoiesis driven by hematopoietic stem cell-like megakaryocyte progenitors. Cell Stem Cell 17, 422–434 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Couldwell, G. & Machlus, K. R. Modulation of megakaryopoiesis and platelet production during inflammation. Thromb. Res. 179, 114–120 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Blüher, M. Metabolically healthy obesity. Endocr. Rev. 41, bnaa004 (2020).

  34. Iacobini, C., Pugliese, G., Blasetti Fantauzzi, C., Federici, M. & Menini, S. Metabolically healthy versus metabolically unhealthy obesity. Metabolism 92, 51–60 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. Xu, S., Jay, A., Brunaldi, K., Huang, N. & Hamilton, J. A. CD36 enhances fatty acid uptake by increasing the rate of intracellular esterification but not transport across the plasma membrane. Biochemistry 52, 7254–7261 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Podrez, E. A. et al. Platelet CD36 links hyperlipidemia, oxidant stress and a prothrombotic phenotype. Nat. Med. 13, 1086–1095 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Meng, O. et al. Loss of Cd36 expression has limited impact on mouse normal hematopoiesis. Blood 140, 11395–11396 (2022).

    Article  Google Scholar 

  38. Xu, X., Zheng, X. & Zhu, F. CD36 gene variants and their clinical relevance: a narrative review. Ann. Blood 6, 34 (2021).

  39. Tomiyama, Y. et al. Identification of the platelet-specific alloantigen, Naka, on platelet membrane glycoprotein IV. Blood 75, 684–687 (1990).

    Article  CAS  PubMed  Google Scholar 

  40. Strassheim, D. et al. Metabolite G-protein coupled receptors in cardio-metabolic diseases. Cells 10, 3347 (2021).

  41. Vijey, P., Posorske, B. & Machlus, K. R. In vitro culture of murine megakaryocytes from fetal liver-derived hematopoietic stem cells. Platelets 29, 583–588 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Heib, T., Gross, C., Muller, M. L., Stegner, D. & Pleines, I. Isolation of murine bone marrow by centrifugation or flushing for the analysis of hematopoietic cells—a comparative study. Platelets 32, 601–607 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Strassel, C. et al. Hirudin and heparin enable efficient megakaryocyte differentiation of mouse bone marrow progenitors. Exp. Cell. Res. 318, 25–32 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Huynh, K. et al. Lipidomic profiling of murine macrophages treated with fatty acids of varying chain length and saturation status. Metabolites 8, 29 (2018).

  45. Weir, J. M. et al. Plasma lipid profiling in a large population-based cohort. J. Lipid Res. 54, 2898–2908 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Huynh, K. et al. High-throughput plasma lipidomics: detailed mapping of the associations with cardiometabolic risk factors. Cell Chem. Biol. 26, 71–84 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Liebisch, G. et al. Update on LIPID MAPS classification, nomenclature, and shorthand notation for MS-derived lipid structures. J. Lipid Res. 61, 1539–1555 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gillespie, M. et al. The reactome pathway knowledgebase 2022. Nucleic Acids Res. 50, D687–D692 (2021).

    Article  PubMed Central  Google Scholar 

  49. French, S. L. et al. High-content, label-free analysis of proplatelet production from megakaryocytes. J. Thromb. Haemost. 18, 2701–2711 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lutz, J. F. & Zarafshani, Z. Efficient construction of therapeutics, bioconjugates, biomaterials and bioactive surfaces using azide–alkyne ‘click’ chemistry. Adv. Drug Deliv. Rev. 60, 958–970 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Hein, C. D., Liu, X. M. & Wang, D. Click chemistry, a powerful tool for pharmaceutical sciences. Pharm. Res. 25, 2216–2230 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kawamoto, T. Use of a new adhesive film for the preparation of multi-purpose fresh-frozen sections from hard tissues, whole-animals, insects and plants. Arch. Histol. Cytol. 66, 123–143 (2003).

    Article  PubMed  Google Scholar 

  53. Watson, S. P., Lowe, G. C., Lordkipanidzé, M. & Morgan, N. V. Genotyping and phenotyping of platelet function disorders. J. Thromb. Haemost. 11, 351–363 (2013).

    Article  PubMed  Google Scholar 

  54. Johnson, B. et al. Whole exome sequencing identifies genetic variants in inherited thrombocytopenia with secondary qualitative function defects. Haematologica 101, 1170–1179 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Thorne, R. F. et al. The integrins α3β1 and α6β1 physically and functionally associate with CD36 in human melanoma cells. J. Biol. Chem. 275, 35264–35275 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Tomlinson, M. et al. Collagen promotes sustained glycoprotein VI signaling in platelets and cell lines. J. Thromb. Haemost. 5, 2274–2283 (2007).

    Article  CAS  PubMed  Google Scholar 

  57. Pekelharing, J. et al. Haematology reference intervals for established and novel parameters in healthy adults. Sysmex Journal International 20, 1–9 (2010).

    Google Scholar 

Download references

Acknowledgements

This work is supported by the National Institute of Diabetes and Digestive and Kidney Diseases (R03DK124746 to K.R.M., R01DK112778 to J.L.) and the National Heart, Lung, and Blood Institute (R01HL151494 to K.R.M., 5T32HL007734 to T.I.H. and R35HL161175 to J.E.I.) (National Institutes of Health); fellowships from the American Society of Hematology (ASH Restart Award to M.N.B.) and the American Heart Association (23POST1011433 to M.N.B.); a Walter Benjamin Fellowship from the German Research Foundation (BE 7766/2-1 to I.C.B.); the Boston Children’s Hospital Surgical Foundation (T.I.H. and M.P.); the Wellcome Trust (218649/Z/19/Z to A.O.K.); the Saudi Arabia Cultural Bureau in London (I.A.); the National Health and Medical Research Council of Australia (APP1194329 to A.J.M. and Investigator Grant 2009965 to P.J.M.); the Fonds de Recherche en Santé du Québec and Canadian Institutes of Health Research (E.B.); and the US Department of Agriculture National Institute of Food and Agriculture (to J.L.).

We would like to thank C. Koo and M. Tomlinson from the University of Birmingham (Birmingham, UK) for assistance with in vitro characterization of the CD36 mutation and B. Nolan (Trinity College Dublin) for referring the patients to the GAPP study. We would also like to thank M. Laplante (University of Laval, Quebec, Canada) and his team for their help with experiments and for being an excellent collaborator. Thank you to J. Cardenas (The University of Texas Health Science Center at Houston) and L. Batista (Washington University in St. Louis) for their critical review of the manuscript, excellent edits and support during the publishing process.

Author information

Authors and Affiliations

Authors

Contributions

M.N.B. conceived the study, performed experiments, collected, interpreted and analyzed data and wrote the manuscript. G.P., I.C.B. and I.A. performed experiments and collected and analyzed data. G.P. prepared all lipidomics samples. A.O.K. provided key intellectual input. E.C. designed and developed image analysis methodologies. D.J.G., D.F., K.G., Z.W. and I.A. performed experiments and analyzed data. T.I.H. and M.P. maintained the CD36 knockout mouse colony and provided experimental and intellectual input. N.V.M. recruited and governed patient ethics, performed experiments, analyzed data and helped write the manuscript. P.K.M. and T.J.C.C. analyzed lipidomic data and prepared corresponding figures. N.A.M. performed lipidomics experiments (with P.J.M.) and uploaded raw data to the metabolights server. P.J.M., J.L. and J.E.I. provided intellectual input and key reagents. E.B. provided reagents, intellectual input and interpreted data. A.J.M. helped conceive the lipidomics study, interpret the data and write the manuscript. K.R.M. conceived and directed the study, analyzed and interpreted data and wrote the manuscript. All authors provided input on and reviewed the manuscript.

Corresponding author

Correspondence to Kellie R. Machlus.

Ethics declarations

Competing interests

J.E.I. has financial interest in and is a founder of Stellular Bio, a biotechnology company focused on making donor-independent platelet-like cells for regenerative medicine. The interests of J.E.I. are managed by Boston Children’s Hospital. All other authors have no conflicts of interest to declare that are relevant to the content of this article.

Peer review

Peer review information

Nature Cardiovascular Research thanks John Hwa, Matthew A. Mitsche and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Megakaryocytes and platelets have unique lipidomic profiles.

Murine bone marrow cell populations were isolated by fluorescence-activated cell sorting and platelets by sequential centrifugation. Lipids were extracted and analyzed using 20-min gradient HPLC and mass spectrometry (see methods for details). (a) Percentage of different lipid classes of indicated murine bone marrow cell populations and autologous platelets in lipidomic analyses. Volcano plots from the different lipid classes between (b) bone marrow extracellular fluid (BMEF) and MEPs. n = 4 and n = 8, respectively, (c) BMEF and immature (CD41+) MKs, n = 4 (d) BMEF and mature (CD41/42+) MKs, n = 4, and (e) platelets and plasma n = 4. Total percentage of all acyl/alkyl composition of (f) PC (g) PI, (h) PE, (i) PS. n = 8 for MEP and 4 for all other cell types, biological replicates. All data are presented as mean +/− SD. MK: megakaryocyte; MEP: MK-erythroid progenitor; PA: phosphatidic acid; PC: phosphatidylcholine; PE: phosphatidylethanolamine; PI: phosphatidylinositol; PS: phosphatidylserine; PG: phosphatidylglycerol.

Source data

Extended Data Fig. 2 Fatty acid uptake and synthesis affected MK differentiation but not mitochondria metabolism.

(a) Fetal liver-derived HSPCs were cultured with TPO and treated with the ACSL inhibitor Triacsin C at indicated doses. CD41+ and CD41/CD42d+ cells were quantified using flow cytometry, n = 5, one-way ANOVA – Dunnett’s test. Data are presented as mean +/− SD. (b) Cytotoxicity assay was performed on day 4 after fetal liver MKs were treated with Triacsin C. Control=cells treated with the vehicle; positive control=cells lysed with TritonX-100. n = 4. Data are presented as mean +/− SEM (c) Fetal liver derived HSPCs were cultured with TPO and treated with the ACC inhibitor PF-05175175 at indicated doses. CD41+ and CD41/CD42d+ cells were quantified using flow cytometry, n = 5, one-way ANOVA – Dunnett’s test. Data are presented as mean +/− SD (d) Cytotoxicity assay was performed day 4 after fetal liver MKs treated with PF-05175175. Control=cells treated with the vehicle; positive control=cells lysed with TritonX-100. n = 4. Data are presented as mean +/− SEM (e) Fetal liver derived HSPCs were cultured with TPO and treated with the FASN inhibitor Cerulenin at indicated doses CD41+ and CD41/CD42d+ cells were quantified using flow cytometry, n = 5, one-way ANOVA – Dunnett’s test. Data are presented as mean +/− SD (f) Cytotoxicity assay was performed day 4 after fetal liver MKs were treated with Cerulenin. Control=cells treated with the vehicle; positive control=cells lysed with TritonX-100, n = 4. Data are presented as mean +/− SEM (g) Representative graph of mitostress assay. MKs were treated with Triacsin C (T4540, Sigma), PF-05175175 (PZ0299, Sigma), Cerulenin (C2389, Sigma) at the indicated concentrations in complete media for 90 min prior the measurement. Oxygen consumption rates were measured in accordance with manufacturer instructions (Agilent/Seahorse Bioscience) n = 5 biological replicates (h) Individual parameters for basal respiration, maximal respiration, and spare respiratory capacity were measured and analyzed. n = 5 biological replicates, one-way ANOVA. Data are presented as mean +/− SD.

Source data

Extended Data Fig. 3 Blood cell counts and characteristics in SFA-enriched high fat diet.

Blood parameters were measured using a Sysmex hematology analyzer, (a) Platelet Distribution Width (PDW), (b) Mean Platelet Volume (MPV), (c) red blood cells, (d) white blood cells, (e) neutrophils; WT n = 12 and Cd36/ n = 16, animals per group, unpaired t-test (f) Platelet lifespan was assessed by flow cytometry measuring the fluorescence-positive platelet population at the indicated time points after injection of biotin-NHS. n = 5 animals per group, unpaired t-test. All data are presented as mean +/− SD.

Source data

Extended Data Fig. 4 Blood cell counts and characteristics in PUFA-enriched high fat diet.

Blood parameters were measured using a Sysmex hematology analyzer, (a) Platelet Distribution Width (PDW), (b) Mean Platelet Volume (MPV), (c) red blood cells; n = 10, unpaired t-test. (d) Platelet lifespan was assessed by flow cytometry measuring the fluorescence-positive platelet population at the indicated time points after injection of biotin-NHS. n = 5, unpaired t-test. Data are presented as mean +/− SD.

Source data

Extended Data Fig. 5 Candidate gene panel analysis.

Schematic of the bioinformatic pipeline workflow used for patient recruitment in the UK-GAPP study.

Extended Data Fig. 6 Mutant CD36 constructs are not trafficked to the cell surface.

WT and mutant CD36 constructs were transfected into both (a) Jurkat T cells and (b) HEK293 cells and surface expression was assessed. Flow cytometry scatter plots indicate that only wildtype CD36 is detected on the cell surface for both cell types. n = 3 biological replicates, unpaired t-test.

Extended Data Fig. 7 Gating strategies for flow cytometry panels.

In vitro studies with bone marrow and fetal liver MKs. Gating strategies of CD41+ and CD41/CD42d+ cells were analyzed using FlowJo. (b) Gating strategies to analyze hematopoietic stem cells and progenitors using FlowJo.

Extended Data Table 1 Thrombopoietin measurements in plasma from mice fed with chow, DIO and PUFA-enriched diets
Extended Data Table 2 Summary of variants included in the UK-GAPP study

Supplementary information

Reporting Summary

Supplementary Table 1

HPLC gradient.

Supplementary Table 2

Mass pairs used for identifying lipids.

Source data

Source Data Fig. 1

Raw data for bulk RNA-seq experiments, detailed normalized data for all lipid species identified in the lipidomics study and Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig./Table 1

Statistical Source Data.

Source Data Extended Data Fig./Table 2

Statistical Source Data.

Source Data Extended Data Fig./Table 3

Statistical Source Data.

Source Data Extended Data Fig./Table 4

Statistical 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

Barrachina, M.N., Pernes, G., Becker, I.C. et al. Efficient megakaryopoiesis and platelet production require phospholipid remodeling and PUFA uptake through CD36. Nat Cardiovasc Res 2, 746–763 (2023). https://doi.org/10.1038/s44161-023-00305-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-023-00305-y

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