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:

Cholesterol modulates the physiological response to nanoparticles by changing the composition of protein corona

Abstract

Nanoparticles (NPs) in biological fluids form a layer of biomolecules known as the protein corona. The protein corona has been shown to determine the biological identity and in vivo fate of NPs, but whether and how metabolites, especially disease-related small molecules, regulate the protein corona and subsequently impact NP fate in vivo is relatively poorly understood. Here we report on the effects of cholesterol on the generation of protein corona and subsequent effects. We find that high levels of cholesterol, as in hypercholesterolemia, result in a protein corona with enriched apolipoproteins and reduced complement proteins by altering the binding affinity of the proteins to the NPs. The cholesterol-mediated protein corona can induce stronger inflammatory responses to NPs in macrophages and promote the cellular uptake of NPs in hepatocytes by enhancing the recognition of lipoprotein receptors when compared with normal protein corona. The result of in vivo biodistribution assays shows that, compared with healthy mice, NPs in hypercholesterolemic mice were more likely to be delivered to the liver, spleen and brain, and less likely to be delivered to the lungs. Our findings reveal that the metabolome profile is an unexploited factor impacting the target efficacy and safety of nanomedicines, providing a way to develop personalized nanomedicines by harnessing disease-related metabolites.

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: Dissecting cholesterol-mediated PC formation.
Fig. 2: Proteomic fingerprints of PC modulated by cholesterol.
Fig. 3: Cho-PC elicited a stronger immune response of macrophages to NPs.
Fig. 4: Cho-PC enhanced cellular internalization of NPs in hepatocytes.
Fig. 5: In vivo biodistribution of NPs in normal and hypercholesterolemic mice.
Fig. 6: The proposed fates of NPs in healthy and hypercholesterolemic individuals.

Similar content being viewed by others

Data availability

All proteomics raw data were deposited to the ProteomeXchange (https://proteomecentral.proteomexchange.org/cgi/GetDataset) with the identifier PXD041073. Source data are provided with this paper. Additional datasets supporting our findings in this study are available from the corresponding authors upon reasonable request.

Code availability

The analysis codes for molecular dynamics simulation and bioinformatic analysis of proteomic data are preserved in the China Academy of Chinese Medical Sciences and are available from the corresponding authors upon reasonable request.

References

  1. Quesada-Gonzalez, D. & Merkoci, A. Nanomaterial-based devices for point-of-care diagnostic applications. Chem. Soc. Rev. 47, 4697–4709 (2018).

    Article  CAS  Google Scholar 

  2. Irvine, D. J. & Dane, E. L. Enhancing cancer immunotherapy with nanomedicine. Nat. Rev. Immunol. 20, 321–334 (2020).

    Article  CAS  Google Scholar 

  3. Stater, E. P., Sonay, A. Y., Hart, C. & Grimm, J. The ancillary effects of nanoparticles and their implications for nanomedicine. Nat. Nanotechnol. 16, 1180–1194 (2021).

    Article  CAS  Google Scholar 

  4. Guerrini, G., Magrì, D., Gioria, S., Medaglini, D. & Calzolai, L. Characterization of nanoparticles-based vaccines for COVID-19. Nat. Nanotechnol. 17, 570–576 (2022).

    Article  CAS  Google Scholar 

  5. Gadekar, V. et al. Nanomedicines accessible in the market for clinical interventions. J. Control. Release 330, 372–397 (2021).

    Article  CAS  Google Scholar 

  6. Thi, T. T. H. et al. Lipid-based nanoparticles in the clinic and clinical trials: from cancer nanomedicine to COVID-19 vaccines. Vaccines 9, 359 (2021).

    Article  CAS  Google Scholar 

  7. Monopoli, M. P., Åberg, C., Salvati, A. & Dawson, K. A. Biomolecular coronas provide the biological identity of nanosized materials. Nat. Nanotechnol. 7, 779–786 (2012).

    Article  CAS  Google Scholar 

  8. Ren, J. et al. Chemical and biophysical signatures of the protein corona in nanomedicine. J. Am. Chem. Soc. 144, 9184–9205 (2022).

    Article  CAS  Google Scholar 

  9. Latreille, P.-L. et al. Scratching the surface of the protein corona: challenging measurements and controversies. ACS Nano 16, 1689–1707 (2022).

    Article  CAS  Google Scholar 

  10. Li, M. et al. Nanoparticle elasticity affects systemic circulation lifetime by modulating adsorption of apolipoprotein A-I in corona formation. Nat. Commun. 13, 4137 (2022).

    Article  CAS  Google Scholar 

  11. Kamaly, N. Nanoparticle protein corona evolution: from biological impact to biomarker discovery. Nanoscale 14, 1606–1620 (2022).

    Article  CAS  Google Scholar 

  12. Ju, Y. et al. Person-specific biomolecular coronas modulate nanoparticle interactions with immune cells in human blood. ACS Nano 14, 15723–15737 (2020).

    Article  CAS  Google Scholar 

  13. Hajipour, M. J., Laurent, S., Aghaie, A., Rezaee, F. & Mahmoudi, M. Personalized protein coronas: a ‘key’ factor at the nano-bio interface. Biomater. Sci. 2, 1210–1221 (2014).

    Article  CAS  Google Scholar 

  14. Shannahan, J. H. et al. From the cover: disease-induced disparities in formation of the nanoparticle-biocorona and the toxicological consequences. Toxicol. Sci. 152, 406–416 (2016).

    Article  CAS  Google Scholar 

  15. Ren, J. et al. Precision nanomedicine development based on specific opsonization of human cancer patient-personalized protein coronas. Nano Lett. 19, 4692–4701 (2019).

    Article  CAS  Google Scholar 

  16. Lazarovits, J. et al. Synthesis of patient-specific nanomaterials. Nano Lett. 19, 116–123 (2019).

    Article  CAS  Google Scholar 

  17. Di Santo, R. et al. Personalized graphene oxide-protein corona in the human plasma of pancreatic cancer patients. Front. Bioeng. Biotechnol. 8, 491 (2020).

    Article  Google Scholar 

  18. Chetwynd, A. J. & Lynch, I. The rise of the nanomaterial metabolite corona, and emergence of the complete corona. Environ. Sci. Nano 7, 1041–1060 (2020).

    Article  CAS  Google Scholar 

  19. Raesch, S. S. et al. Proteomic and lipidomic analysis of nanoparticle corona upon contact with lung surfactant reveals differences in protein, but not lipid composition. ACS Nano 9, 11872–11885 (2015).

    Article  CAS  Google Scholar 

  20. Braccia, C. et al. The lipid composition of few layers graphene and graphene oxide biomolecular corona. Carbon 185, 591–598 (2021).

    Article  CAS  Google Scholar 

  21. Liu, K., Salvati, A. & Sabirsh, A. Physiology, pathology and the biomolecular corona: the confounding factors in nanomedicine design. Nanoscale 14, 2136–2154 (2022).

    Article  CAS  Google Scholar 

  22. Kobos, L. M. et al. An integrative proteomic/lipidomic analysis of the gold nanoparticle biocorona in healthy and obese conditions. Appl. Vitr. Toxicol. 5, 150–166 (2019).

    Article  CAS  Google Scholar 

  23. Lima, T., Bernfur, K., Vilanova, M. & Cedervall, T. Understanding the lipid and protein corona formation on different sized polymeric nanoparticles. Sci. Rep. 10, 1129 (2020).

    Article  CAS  Google Scholar 

  24. Tavakol, M. et al. Disease-related metabolites affect protein-nanoparticle interactions. Nanoscale 10, 7108–7115 (2018).

    Article  CAS  Google Scholar 

  25. Luo, J., Yang, H. & Song, B. L. Mechanisms and regulation of cholesterol homeostasis. Nat. Rev. Mol. Cell Biol. 21, 225–245 (2020).

    Article  CAS  Google Scholar 

  26. Civeira, F., Arca, M., Cenarro, A. & Hegele, R. A. A mechanism-based operational definition and classification of hypercholesterolemia. J. Clin. Lipidol. 16, 813–821 (2022).

    Article  Google Scholar 

  27. Kim, S. H. et al. Understanding the biomolecular coronas of high-density lipoproteins on pegylated Au nanoparticles: implication for lipid corona formation in the blood. ACS Appl. Nano Mater. 5, 2018–2028 (2022).

    Article  CAS  Google Scholar 

  28. Kim, H., Kumar, S., Kang, D. W., Jo, H. & Park, J. H. Affinity-driven design of cargo-switching nanoparticles to leverage a cholesterol-rich microenvironment for atherosclerosis therapy. ACS Nano 14, 6519–6531 (2020).

    Article  CAS  Google Scholar 

  29. Fu, Q., Yu, L., Wang, Y., Li, P. & Song, J. Biomarker-responsive nanosystems for chronic disease theranostics. Adv. Funct. Mater. 33, 2206300 (2023).

    Article  CAS  Google Scholar 

  30. Kim, K. R., Kim, J., Back, J. H., Lee, J. E. & Ahn, D. R. Cholesterol-mediated seeding of protein corona on dna nanostructures for targeted delivery of oligonucleotide therapeutics to treat liver fibrosis. ACS Nano 16, 7331–7343 (2022).

    Article  CAS  Google Scholar 

  31. Dreaden, E. C., Alkilany, A. M., Huang, X., Murphy, C. J. & El-Sayed, M. A. The golden age: gold nanoparticles for biomedicine. Chem. Soc. Rev. 41, 2740–2779 (2012).

    Article  CAS  Google Scholar 

  32. Kim, S. E. et al. Ultrasmall nanoparticles induce ferroptosis in nutrient-deprived cancer cells and suppress tumour growth. Nat. Nanotechnol. 11, 977–985 (2016).

    Article  CAS  Google Scholar 

  33. Ke, P. C., Lin, S., Parak, W. J., Davis, T. P. & Caruso, F. A decade of the protein corona. ACS Nano 11, 11773–11776 (2017).

    Article  CAS  Google Scholar 

  34. Xiao, Q. et al. The effects of protein corona on in vivo fate of nanocarriers. Adv. Drug Deliv. Rev. 186, 114356 (2022).

    Article  CAS  Google Scholar 

  35. Blanco, E., Shen, H. & Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 33, 941–951 (2015).

    Article  CAS  Google Scholar 

  36. Macia, E. et al. Dynasore, a cell-permeable inhibitor of dynamin. Dev. Cell 10, 839–850 (2006).

    Article  CAS  Google Scholar 

  37. Francia, V. et al. Corona composition can affect the mechanisms cells use to internalize nanoparticles. ACS Nano 13, 11107–11121 (2019).

    Article  CAS  Google Scholar 

  38. Lara, S. et al. Identification of receptor binding to the biomolecular corona of nanoparticles. ACS Nano 11, 1884–1893 (2017).

    Article  CAS  Google Scholar 

  39. Ngo, W. et al. Identifying cell receptors for the nanoparticle protein corona using genome screens. Nat. Chem. Biol. 18, 1023–1031 (2022).

    Article  CAS  Google Scholar 

  40. Benetti, F., Bregoli, L., Olivato, I. & Sabbioni, E. Effects of metal (loid)-based nanomaterials on essential element homeostasis: the central role of nanometallomics for nanotoxicology. Metallomics 6, 729–747 (2014).

    Article  CAS  Google Scholar 

  41. Ashkarran, A. A. et al. Measurements of heterogeneity in proteomics analysis of the nanoparticle protein corona across core facilities. Nat. Commun. 13, 6610 (2022).

    Article  CAS  Google Scholar 

  42. Sheibani, S. et al. Nanoscale characterization of the biomolecular corona by cryo-electron microscopy, cryo-electron tomography, and image simulation. Nat. Commun. 12, 573 (2021).

    Article  CAS  Google Scholar 

  43. Shevchenko, A., Tomas, H., Havlis, J., Olsen, J. V. & Mann, M. In-gel digestion for mass spectrometric characterization of proteins and proteomes. Nat. Protoc. 1, 2856–2860 (2006).

    Article  CAS  Google Scholar 

  44. Cao, Z. T. et al. Protein binding affinity of polymeric nanoparticles as a direct indicator of their pharmacokinetics. ACS Nano 14, 3563–3575 (2020).

    Article  CAS  Google Scholar 

  45. Hess, B. P-LINCS: a parallel linear constraint solver for molecular simulation. J. Chem. Theory Comput. 4, 116–122 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The work was supported by grants from the National Key Research and Development Program of China (2020YFA0908000 and 2022YFC2303600); the Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (no. ZYYCXTD-C-202002); the National Natural Science Foundation of China (32201177, 32000026, 82141001, 82274182 and 82074098); the CACMS Innovation Fund (CI2021A05101 and CI2021A05104); the Scientific and Technological Innovation Project of China Academy of Chinese Medical Sciences (CI2021B014); the Science and Technology Foundation of Shenzhen (JCYJ20210324115800001); the Science and Technology Foundation of Shenzhen (Shenzhen Clinical Medical Research Center for Geriatric Diseases); the National Key R&D Program of China Key projects for international cooperation on science, technology and innovation (2020YFE0205100); the Fundamental Research Funds for the Central Public Welfare Research Institutes (ZZ14-YQ-061, ZZ16-ND-10–24, ZZ14-YQ-057, ZZ14-YQ-050, ZZ14-YQ-051, ZZ14-FL-002, ZZ14-ND-010 and ZZ15-ND-10); GuangDong Basic and Applied Basic Research Foundation (2021A1515012164), Shenzhen Key Laboratory of Kidney Diseases (ZDSYS201504301616234); and Shenzhen Governmental Sustainable Development Fund (KCXFZ20201221173612034).

Author information

Authors and Affiliations

Authors

Contributions

J.W., C.X. and H.T. conceived the project and supervised the study. H.T. and Y. Zhang designed and performed all experiments with the help from the other authors. T.Y., C.W., L.Q. and J.Z. assisted in the collection and analysis of proteomic data. Y. Zhu, C.W. and Y.K.W. assisted in the molecular dynamics simulation. J.L., Y.S. and L.Z assisted in the animal experiment and inductively coupled plasma mass spectrometry measurement. Y.L. and H.W. guided and assisted in the experiment design. H.T., Y. Zhang, C.X. and J.W. analysed data and wrote the paper. J.W., C.X. and Y.K.W. edited the paper. All authors approved the paper.

Corresponding authors

Correspondence to Chengchao Xu or Jigang Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Nanotechnology thanks Andy Chetwynd, Morteza Mahmoudi 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 Characterization of cholesterol-mediated protein corona on NPs.

(a) Coomassie-stained SDS-PAGE gel of PC on SNPs under different concentrations of cholesterol. (b) The total protein amounts of PC on SNPs under different concentrations of cholesterol. The data were presented as mean ± standard deviation with n = 3 (biologically independent experiments). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (c) Densitometric analysis of Extended Data Fig. 1a. (d) Densitometric analysis of Fig. 1a. (e) Coomassie-stained SDS-PAGE gel of human serum samples with different concentrations of cholesterol.

Source data

Extended Data Fig. 2 Characterization of NPs-Nor-PC and NPs-Cho-PC.

(a) Cryo-TEM images of NPs, NPs-Nor-PC, and NPs-Cho-PC. Cryo-TEM imaging showed that cholesterol did not affect the possible protein contamination in the background. (b) Hydrodynamic (HD) size distribution AuNPs, AuNPs-Nor-PC, and AuNPs-Cho-PC. (c) HD size distribution of SNPs, SNPs-Nor-PC, and SNPs-Cho-PC. (d) Summary of physicochemical parameters including TEM size, HD, polydispersity index (PDI), and zeta potential of naked NPs, NPs-Nor-PC, and NPs-Cho-PC for AuNPs and SNPs. (e) The loading amount of cholesterol on SNPs under different concentrations of cholesterol. The data were presented as mean ± standard deviation with n = 3 (biologically independent experiments).

Source data

Extended Data Fig. 3 Schematic illustration for the mechanistic formation of Cho-PC and proteomic fingerprints identification.

(a) Schematic process of PC on SNPs under different conditions for elucidating the mechanism of Cho-PC formation. PC-I refers to the normal PC formed in FBS. PC-II refers to the PC formed in FBS after SNPs were pre-incubated with the cholesterol solution. PC-III refers to the final PC formed in FBS containing cholesterol after SNPs were pre-incubated with FBS. PC-IV refers to the Cho-PC formed in FBS containing cholesterol. (b) Schematic diagram of identification and quantification of Cho-PC and Nor-PC at the proteomic level.

Extended Data Fig. 4 Proteomic fingerprints identification and quantification of Cho-PC.

(a-c) Comparison of the number of PC identified under different concentrations of cholesterol according to their functional classification (a), isoelectric point (b), and molecular weight (c). (d-f) The biological function (d), cell component (e), and molecular function (f) category of the significantly changed proteins in Cho-PC compared with Nor-PC according to the GO classification. (g) The enriched KEGG pathway of the significantly changed proteins in Cho-PC compared with Nor-PC. (h) Heat map of the top 25 abundance proteins in Nor-PC and Cho-PC with the clustering analysis. (i-j) Relative abundances of the identified apolipoprotein proteins (i) and complement proteins (j) in Nor-PC and Cho-PC. The data were presented as mean ± standard deviation with n = 3 (biologically independent experiments).

Source data

Extended Data Fig. 5 Proteomic analysis of Cho-PC in human serum and adsorption of LDL and HDL on NPs.

(a)The heatmap of apolipoproteins and complement proteins identified in PC when bare SNPs and cholesterol-precoated SNPs were incubated with the normal FBS. Cholesterol precoated SNPs were prepared by incubating bare SNPs in 5.1 mM cholesterol solution and subsequent three PBS washing. (b-e) The heatmaps (b, d) and relative protein abundance (c, e) of apolipoproteins (b, c) and complement proteins (d, e) identified in neat serum and protein corona on SNPs in human serum with different concentrations of cholesterol. (f-g) The amount of LDL (f) and HDL (g) adsorbed on SNPs by measuring the concentrations in supernatants before and after SNPs addition. The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments).

Source data

Extended Data Fig. 6 Circular dichroism analysis of the interactions of proteins with SNPs in the absence or presence of cholesterol.

(a-h) Circular dichroism (a, c, e, g) and secondary structure (b, d, f, h) of APOE (a-b), C1Q (c-d), LDL (e-f) and HDL (h-g) in the absence and presence of cholesterol or SNPs. The conformation of APOE and C1Q changed after interacting with SNPs. Even though cholesterol itself did not induce conformational changes, it further enhanced the conformational change of APOE elicited by SNPs. The conformation of LDL and HDL could be altered by both cholesterol and SNPs, and the change became more distinct in the presence of both cholesterol and SNPs.

Source data

Extended Data Fig. 7 The effects of Nor-PC and Cho-PC on cytotoxicity and immune response of macrophages to NPs.

(a-b) Cytotoxicity of AuNPs (a) and SNPs (b) with various coatings on Raw264.7 cells at 24 h. The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (c-d) Volcano plots of the protein expression change for the Nor-PC group (c) and Nor-PC group (d) versus the control group. (e) The overlap of the significantly changed proteins in the Cho-PC group and Nor-PC group versus the control group. (f) The enriched biological pathway of down-regulated proteins in the Nor-PC group and Cho-PC group by GO analysis.

Source data

Extended Data Fig. 8 The effects of Cho-PC on cellular internalization of NPs in hepatocytes.

(a) Cell viability assessment of AuNPs, AuNPs-Nor-PC, and AuNPs-Cho-PC on HepG2 cells. The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (b) Cell viability assessment of SNPs, SNPs-Nor-PC, and SNPs-Cho-PC on HepG2 cells. The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (c) Uptake kinetics of SNPs-Nor-PC and SNPs-Cho-PC in the media with or without FBS. (d-e) Cellular uptake of 100 µg ml−1 of FITC labelled SNPs in HepG2 cells under different concentrations of cholesterol in the medium with FBS quantified by flow cytometry. The average and standard deviation represented three replicates of the median cell fluorescence intensities obtained. The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (f-g) Cellular uptake of 100 µg ml−1 of bare SNPs, SNPs-Nor-PC and SNPs-Cho-PC in HepG2 cells in serum-free medium in the presence and absence of cholesterol. The average and standard deviation represented three replicates of the median cell fluorescence intensities obtained. The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (h-m) The uptake kinetics of SNPs-Nor-PC and SNPs-Cho-PC in the presence or absence of 10 μg/mL chlorpromazine (h), 2.5 mg/mL methyl-β-cyclodextrin (i), 25 μg/mL dynasore (j), 100 μM EIPA (k), 2.5 μg/mL cytochalasin D (l), and 5 μM nocodazole (m). The data are presented as mean ± standard deviation with n = 3 (biologically independent experiments).

Source data

Extended Data Fig. 9 Characterization of Cho-PC in normal and hypercholesterolemia mice.

(a-b) SDS-PAGE characterization of PC formed on AuNPs immersed in plasmas (a) and neat plasmas (b) from the normal and hypercholesterolemia mice. (c-d) SDS-PAGE characterization of PC formed on AuNPs immersed in serums (c) and neat serums (d) from the normal and hypercholesterolemia mice. (e) The heatmap of apolipoproteins and complement proteins identified in neat serum and protein corona on AuNPs after incubation with the serum from normal and hypercholesterolemic mice. (f) The heatmap of apolipoproteins and complement proteins identified in PC when SNPs were incubated with the serums from normal and hypercholesterolemic mice.

Source data

Extended Data Fig. 10 In vivo biodistribution of NPs in normal and hypercholesterolemia mice.

(a-b) The concentrations of Au in blood and tissues from the normal and hyperlipidemia mice at 1 h (a) and 24 h (b) after intravenous injection of AuNPs at doses of 0.1 g per kg body weight. The data are presented as mean ± standard deviation with n = 5 (biologically independent mice). Statistical significance was tested with a two-tailed, unpaired Student’s t-test. (c, e) Tissue distribution estimated by the fluorescence intensity at 1 h (c) and 12 h (e) after normal and hypercholesterolemic mice were intravenously injected with Cy3-labelled SNPs at doses of 20 mg per kg body weight. The data are presented as mean ± standard deviation with n = 4 (biologically independent mice). Statistical significance was tested with a two-tailed, unpaired Student’s t test. (d, f) Ex vivo fluorescence images of major organs showing the distribution of SNPs in normal and hypercholesterolemic mice at 1 h (d) and 12 h (f). (Bl, blood; Lu, lungs; St, stomach; Te, testis; Li, liver; Sp, spleen; Bo, bone; Br, brain; Ki, kidneys; He, heart; In, intestinal).

Source data

Supplementary information

Supplementary Information

Supplementary Methods, Figs. 1–7, Table 1 and References.

Reporting Summary

Supplementary Video 1

Molecular dynamics trajectory of apolipoprotein E with SNPs in the absence of cholesterol.

Supplementary Video 2

Molecular dynamics trajectory of apolipoprotein E with SNPs in the presence of cholesterol.

Source data

Source Data Fig. 1

Numerical source data and unprocessed gels.

Source Data Fig. 2

Numerical source data and unprocessed western blots.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Numerical source data.

Source Data Extended Data Fig. 1

Numerical source data.

Source Data Extended Data Fig. 2

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 5

Numerical source data.

Source Data Extended Data Fig. 6

Numerical source data.

Source Data Extended Data Fig. 7

Numerical source data.

Source Data Extended Data Fig. 8

Numerical source data.

Source Data Extended Data Fig. 9

Unprocessed gels.

Source Data Extended Data Fig. 10

Numerical 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

Tang, H., Zhang, Y., Yang, T. et al. Cholesterol modulates the physiological response to nanoparticles by changing the composition of protein corona. Nat. Nanotechnol. 18, 1067–1077 (2023). https://doi.org/10.1038/s41565-023-01455-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-023-01455-7

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