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:

Hepatic nonvesicular cholesterol transport is critical for systemic lipid homeostasis

Abstract

In cell models, changes in the ‘accessible’ pool of plasma membrane (PM) cholesterol are linked with the regulation of endoplasmic reticulum sterol synthesis and metabolism by the Aster family of nonvesicular transporters; however, the relevance of such nonvesicular transport mechanisms for lipid homeostasis in vivo has not been defined. Here we reveal two physiological contexts that generate accessible PM cholesterol and engage the Aster pathway in the liver: fasting and reverse cholesterol transport. During fasting, adipose-tissue-derived fatty acids activate hepatocyte sphingomyelinase to liberate sequestered PM cholesterol. Aster-dependent cholesterol transport during fasting facilitates cholesteryl ester formation, cholesterol movement into bile and very low-density lipoprotein production. During reverse cholesterol transport, high-density lipoprotein delivers excess cholesterol to the hepatocyte PM through scavenger receptor class B member 1. Loss of hepatic Asters impairs cholesterol movement into feces, raises plasma cholesterol levels and causes cholesterol accumulation in peripheral tissues. These results reveal fundamental mechanisms by which Aster cholesterol flux contributes to hepatic and systemic lipid homeostasis.

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: Asters transport accessible PM cholesterol in hepatocytes.
Fig. 2: Asters facilitate hepatocyte PM to ER cholesterol transport during fasting.
Fig. 3: Adipose-derived fatty acids liberate sequestered PM cholesterol during fasting.
Fig. 4: Fasting induces Smpd3 expression to regulate cholesterol metabolism and VLDL production.
Fig. 5: Aster-C is a direct target gene of FXR in mouse liver.
Fig. 6: Hepatic Aster deficiency impairs transport of lipoprotein-derived cholesterol and RCT.
Fig. 7: Asters are required for storage of dietary cholesterol as CEs in hepatocytes.
Fig. 8: Hepatic Aster deficiency disrupts systemic cholesterol homeostasis.

Similar content being viewed by others

Data availability

The RNA-seq dataset generated for this paper has been deposited to the National Center for Biotechnology Information and is available at accession no. GSE206278. For RNA-seq analysis, trimmed FASTQ files were aligned to GRCm38/mm10 using STAR40. Source data are provided with this paper.

References

  1. Lange, Y., Swaisgood, M. H., Ramos, B. V. & Steck, T. L. Plasma membranes contain half the phospholipid and 90% of the cholesterol and sphingomyelin in cultured human fibroblasts. J. Biol. Chem. 264, 3786–3793 (1989).

    Article  CAS  PubMed  Google Scholar 

  2. Das, A., Brown, M. S., Anderson, D. D., Goldstein, J. L. & Radhakrishnan, A. Three pools of plasma membrane cholesterol and their relation to cholesterol homeostasis. eLife 3, e02882 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Radhakrishnan, A., Goldstein, J. L., McDonald, J. G. & Brown, M. S. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: a delicate balance. Cell Metab. 8, 512–521 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Anderson, R. A. et al. Identification of a form of acyl-CoA: cholesterol acyltransferase specific to liver and intestine in nonhuman primates. J. Biol. Chem. 273, 26747–26754 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Pikuleva, I. A. Cholesterol-metabolizing cytochromes P450. Drug Metab. Dispos. 34, 513–P520 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Sandhu, J. et al. Aster proteins facilitate nonvesicular plasma membrane to ER cholesterol transport in mammalian cells. Cell 175, 514–529 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Naito, T. et al. Movement of accessible plasma membrane cholesterol by GRAMD1 lipid transfer protein complex. eLife 8, e51401 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xiao, X. et al. Selective Aster inhibitors distinguish vesicular and nonvesicular sterol transport mechanisms. Proc. Natl Acad. Sci. USA 118, e2024149118 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Trinh, M. N. et al. Interplay between Asters/GRAMD1s and phosphatidylserine in intermembrane transport of LDL cholesterol. Proc. Natl Acad. Sci. USA 119, e2120411119 (2022).

  10. Ferrari, A. et al. Aster proteins regulate the accessible cholesterol pool in the plasma membrane. Mol. Cell. Biol. 40, e00255-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Kennelly, J. P. & Tontonoz, P. Cholesterol transport to the endoplasmic reticulum. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a041263 (2022).

  12. Edwards, P. A., Muroya, H. & Gould, R. G. In vivo demonstration of the circadian rhythm of cholesterol biosynthesis inthe liver and intestine of the rat. J. Lipid Res. 13, 396–401 (1972).

    Article  CAS  PubMed  Google Scholar 

  13. Horton, J. D., Bashmakov, Y., Shimomura, I. & Shimano, H. Regulation of sterol regulatory element binding proteins in livers of fasted and refed mice. Proc. Natl Acad. Sci. USA 95, 5987–5992 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tall, A. An overview of reverse cholesterol transport. Atherosclerosis 109, 337 (1994).

    Article  Google Scholar 

  15. Acton, S. et al. Identification of scavenger receptor SR-BI as a high-density lipoprotein receptor. Science 271, 518–520 (1996).

    Article  CAS  PubMed  Google Scholar 

  16. Zhang, Y. et al. Identification of novel pathways that control farnesoid X receptor-mediated hypocholesterolemia. J. Biol. Chem. 285, 3035–3043 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Schaum, N. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  PubMed Central  Google Scholar 

  18. Gay, A., Rye, D. & Radhakrishnan, A. Switch-like responses of two cholesterol sensors do not require protein oligomerization in membranes. Biophys. J. 108, 1459–1469 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Worgall, T. S., Johnson, R. A., Seo, T., Gierens, H. & Deckelbaum, R. J. Unsaturated fatty acid-mediated decreases in sterol regulatory element-mediated gene transcription are linked to cellular sphingolipid metabolism. J. Biol. Chem. 277, 3878–3885 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Simcox, J. et al. Global analysis of plasma lipids identifies liver-derived acylcarnitines as a fuel source for brown fat thermogenesis. Cell Metab. 26, 509–522 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Insausti-Urkia, N., Solsona-Vilarrasa, E., Garcia-Ruiz, C. & Fernandez-Checa, J. C. Sphingomyelinases and liver disease. Biomolecules 10, 1497 (2020).

  22. Thomas, A. M. et al. Genome‐wide tissue‐specific farnesoid X receptor binding in mouse liver and intestine. Hepatology 51, 1410–1419 (2010).

    Article  CAS  PubMed  Google Scholar 

  23. Redgrave, T. G., Roberts, D. C. K. & West, C. E. Separation of plasma lipoproteins by density-gradient ultracentrifugation. Anal. Biochem. 65, 42–49 (1975).

    Article  CAS  PubMed  Google Scholar 

  24. Brown, M. S. & Goldstein, J. L. A receptor-mediated pathway for cholesterol homeostasis. Science 232, 34–47 (1986).

    Article  CAS  PubMed  Google Scholar 

  25. Tabas, I., Rosoff, W. J. & Boykow, G. C. Acyl coenzyme A: cholesterol acyl transferase in macrophages utilizes a cellular pool of cholesterol oxidase-accessible cholesterol as substrate. J. Biol. Chem. 263, 1266–1272 (1988).

    Article  CAS  PubMed  Google Scholar 

  26. Ji, Y. et al. Hepatic scavenger receptor BI promotes rapid clearance of high-density lipoprotein free cholesterol and its transport into bile. J. Biol. Chem. 274, 33398–33402 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Feng, Y. et al. Hepatocyte-specific ABCA1 transfer increases HDL cholesterol but impairs HDL function and accelerates atherosclerosis. Cardiovasc. Res. 88, 376–385 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Singaraja, R. R. et al. Human ABCA1 BAC transgenic mice show increased high-density lipoprotein cholesterol and ApoAI-dependent efflux stimulated by an internal promoter containing liver X receptor response elements in intron 1. J. Biol. Chem. 276, 33969–33979 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Basso, F. et al. Role of the hepatic ABCA1 transporter in modulating intrahepatic cholesterol and plasma HDL cholesterol concentrations. J. Lipid Res. 44, 296–302 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Timmins, J. M. et al. Targeted inactivation of hepatic Abca1 causes profound hypoalphalipoproteinemia and kidney hypercatabolism of apoA-I. J. Clin. Invest. 115, 1333–1342 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Rigotti, A. et al. A targeted mutation in the murine gene encoding the high-density lipoprotein (HDL) receptor scavenger receptor class B type I reveals its key role in HDL metabolism. Proc. Natl Acad. Sci. USA 94, 12610–12615 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lu, X.-Y. et al. Feeding induces cholesterol biosynthesis via the mTORC1–USP20–HMGCR axis. Nature 588, 479–484 (2020).

    Article  CAS  PubMed  Google Scholar 

  33. Yu, L. et al. Expression of ABCG5 and ABCG8 is required for regulation of biliary cholesterol secretion. J. Biol. Chem. 280, 8742–8747 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Kosters, A. et al. The mechanism of ABCG5/ABCG8 in biliary cholesterol secretion in mice 1. J. Lipid Res. 47, 1959–1966 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Clifford, B. L. et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 33, 1671–1684 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sinal, C. J. et al. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102, 731–744 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. de Aguiar Vallim, T. Q. et al. MAFG is a transcriptional repressor of bile acid synthesis and metabolism. Cell Metab. 21, 298–311 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Tarling, E. J. et al. RNA-binding protein ZFP36L1 maintains posttranscriptional regulation of bile acid metabolism. J. Clin. Invest. 127, 3741–3754 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  42. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

  44. Folch, J., Lees, M. & Stanley, G. H. S. A simple method for the isolation and purification of total lipids from animal tissues. J. Biol. Chem. 226, 497–509 (1957).

    Article  CAS  PubMed  Google Scholar 

  45. Hsieh, W.-Y. et al. Toll-like receptors induce signal-specific reprogramming of the macrophage lipidome. Cell Metab. 32, 128–143 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Su, B. et al. A DMS shotgun lipidomics workflow application to facilitate high-throughput, comprehensive lipidomics. J. Am. Soc. Mass Spectrom. 32, 2655–2663 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lyu, K. et al. A membrane-bound diacylglycerol species induces PKCϵ-mediated hepatic insulin resistance. Cell Metab. 32, 654–664 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Endapally, S., Infante, R. E. & Radhakrishnan, A. Monitoring and modulating intracellular cholesterol trafficking using ALOD4, a cholesterol-binding protein. Methods Mol. Biol. 1949, 153–163 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhou, Q. D. et al. Interferon-mediated reprogramming of membrane cholesterol to evade bacterial toxins. Nat. Immunol. 21, 746–755 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Robichaud, J. C., van der Veen, J. N., Yao, Z., Trigatti, B. & Vance, D. E. Hepatic uptake and metabolism of phosphatidylcholine associated with high-density lipoproteins. Biochim. Biophys. Acta 1790, 538–551 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Terpstra, A. H., Nicolosi, R. J. & Herbert, P. N. In vitro incorporation of radiolabeled cholesteryl esters into high and low-density lipoproteins. J. Lipid Res. 30, 1663–1671 (1989).

    Article  CAS  PubMed  Google Scholar 

  52. Jayadev, S., Linardic, C. M. & Hannun, Y. A. Identification of arachidonic acid as a mediator of sphingomyelin hydrolysis in response to tumor necrosis factor α. J. Biol. Chem. 269, 5757–5763 (1994).

    Article  CAS  PubMed  Google Scholar 

  53. Low, H., Hoang, A. & Sviridov, D. Cholesterol efflux assay. J. Vis. Exp. https://doi.org/10.3791/3810 (2012).

  54. Zabalawi, M. et al. Inflammation and skin cholesterol in LDLr−/−, apoA-I−/− mice: link between cholesterol homeostasis and self-tolerance? J. Lipid Res. 48, 52–65 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all members of the Tontonoz, Tarling-Vallim, Edwards, Villanueva, Young and Bensinger laboratories at UCLA for useful advice and discussions and for sharing reagents and resources. We thank K. Williams, G. Su and staff at UCLA Lipidomics core for the lipidomics analysis. Confocal microscopy was performed at the California NanoSystems Institute of Advanced Light Microscopy/Spectroscopy Facility. We thank the Vector Core of the University of Michigan for AAV packaging. We thank J. Smothers and A. Radhakrishnan for the helpful suggestions about the ALOD4 protein purification. This work was supported by NIH grant R01 DK126779 and Fondation Leducq Transatlantic Network of Excellence (19CVD04). X.X. was supported by AHA Postdoctoral Fellowship (18POST34030388). J.P.K. is supported by AHA Postdoctoral Fellowship (903306). A.F. was funded by Ermenegildo Zegna Founder’s Scholarship (2017) and by American Diabetes Association postdoctoral fellowship (1-19-PDF-043-RA). Y.G. is supported by Damon Runyon Cancer Research Foundation and Mark Foundation postdoctoral fellowship (DRG2424-21). R.T.N. was supported by a T32GM008042 grant to the UCLA-Caltech Medical Scientist Training Program. A.N. was supported by the NIDDK of the National Institutes of Health under Award Number T32DK007180.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization was carried out by X.X., J.P.K. and P.T. Methodology was the responsibility of X.X., J.P.K., B.L.C., Y.G., K.Q., J.S., A.N., R.T.N. and M.S.L. Investigation was conducted by X.X., J.P.K., A.F., B.L.C., E.W., Y.G., K.Q., J.S., K.E.J, M.C.B., A.N., R.T.N., M.S.L., S.Z. and T.W. Writing was carried out by X.X., J.P.K. and P.T. Funding was acquired by P.T. Resources were the responsibility of S.G.Y., S.J.B., C.J.V., T.Q.d.A.V. and P.T. Supervision was carried out by P.T.

Corresponding author

Correspondence to Peter Tontonoz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks David Cohen Andrew Brown and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Isabella Samuelson, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 Generation of Aster-C 3xHA KI and Asters hepatocyte specific KO mice.

a, Quantitative PCR of Asters expression in mouse liver (n = 6). b, Evaluation of 3xHA-Aster-C KI mice. Genotyping results (upper) and anti-HA tag immunoblot (bottom) for 3xHA-Aster-C KI mice. The genotyping result is representative of at least 50 similar results. The anti-HA-Aster-C western blot was repeated independently in Fig. 5e. c, Strategy for generating Gramd1a (Aster-A) and Gramd1c (Aster-C) Flox/Flox (F/F) mice. Coding exons are depicted in black. Exons that correspond to the GRAM domain, ASTER domain, and transmembrane (TM) domain are depicted in green, blue, and red respectively. Scale bar represents 1 kb. d, Genotyping results for L-A KO, L-C KO and L-A/C KO mice. The genotyping results are representative of at least 100 similar results per condition. (e, f and g) Expression levels of the indicated genes in liver from F/F control and L-A KO mice (e, n = 8/11); L-C KO mice (f, n = 5/6); L-A/C KO mice (g, n = 12/6). h, Gramd1a, Gramd1b and Gramd1c expression level in the testis of F/F control and L-A/C KO mice (n = 12/6). All data are presented as mean ± SEM. P values were determined by two-sided Student’s t-test with Benjamini, Krieger and Yekutieli correction for multiple comparisons (e, f, g and h).

Source data

Extended Data Fig. 2 Fasting stimulates hepatic PM–ER cholesterol transport.

a, Gross appearance of livers from mice fasted for 4- or 16-h. b, Hepatic triglycerides in mice fasted for 4- or 16-h (n = 3/3). c, Hepatic CE in mice fasted for 4- or 16-h (n = 3/3). d, Significantly upregulated pathways in the livers of mice fasted for 16-h compared to 4-h according to pathway analysis of RNA Sequencing data. e, Significantly downregulated pathways in the livers of mice fasted for 16-h compared to 4-h according to pathway analysis of RNA Sequencing data. f, Hepatic mRNA expression of SREBP-2 pathway targets from the livers of mice fasted for 4- or 16-h (n = 3/3). g, Quality control of plasma membrane isolation from the mouse liver. Cadherin: PM maker; Calnexin: ER maker; ATGL: lipid droplet (LD) maker; Actin: cytoskeleton maker. This analysis was completed once as the organelle isolation method has been previously validated (further method details are in the Methods section). h, Free cholesterol analysis from purified PMs of wild-type mice (n = 3/3). i, TLC analysis of free cholesterol (FC), sphingomyelin (SM) and phosphatidylcholine (PC) from the livers of mice fasted for either 4 or 16 h. j, PM total lipids as measured by mass-spec from livers of F/F control and L-A/C KO after 4- and 16-h fasting (n = 3/3). All data are presented as mean ± SEM. P values were determined by two-sided Student’s t-test (b, c and h), or two-sided Student’s t-test with Benjamini, Krieger and Yekutieli correction (f).

Source data

Extended Data Fig. 3 Aster-mediated cholesterol transport determines the size of the accessible PM cholesterol pool.

a, PM total lipids from mouse liver after 4- and 16-h fasting as determined by mass spec (n = 3/3). b, Immunoblot analysis of ALOD4 binding in F/F control and L-A/C KO primary hepatocytes after treatment with vehicle, nSmase (100 mU/ml) and GW4869 (10 μM) for 1-h. Actin was used as a loading control. c, Expression levels of the indicated genes in primary hepatocyte that had been cultured in Maintenance medium and Ro 48-8071 (1μM) overnight before being treated with or without oleic acid (30μM) for 6-h (n = 3). d, Immunoblot analysis of ALOD4 binding in primary hepatocytes after treatment with vehicle or indicated concentration of glucagon, acetoacetate (Ac-Ac), beta-hydroxybutyrate (BHB) or MβCD-cholesterol (35 µM) for 1-h. Calnexin was used as a loading control. e, Immunoblot analysis of ALOD4 binding in primary hepatocytes after treatment with vehicle or insulin (300 nM), glucose (50 mM) and both for 1-h. Calnexin was used as a loading control. All data are presented as mean ± SEM.

Source data

Extended Data Fig. 4 Asters transport lipoprotein-derived cholesterol from the PM to ER in hepatocytes.

a. Immunoblot analysis of ALOD4 binding in control or Sr-b1 knockdown HAECs after treatment with vehicle or HDL (400 ug/ml) for 1-h. Calnexin was used as a sample processing control. b, [14C] counts in liver unesterified cholesterol of mice from Fig. 6c (n = 10/6). c, mRNA expression levels of the indicated genes in the livers of mice from Fig. 6c (n = 10/6). d, [14C] counts in unesterified cholesterol in the livers of mice from Fig. 6g (n = 7/9). e, mRNA expression levels of the indicated genes in the livers of mice from Fig. 6g (n = 7/9). f, The rate of [14C] lipoprotein clearance from the circulation (n = 6/5), related to Figs. 6j6m. Data are represented as mean ± SEM with individual animals noted as dots. *p < 0.05. g. Fecal cholesterol analysis of F/F and L-A/C KO mice fed a cholesterol-free diet for 48 hours (n = 7/7). h, Growth curves for F/F and L-A/C KO mice fed a Western diet from 8 weeks of age (n = 8/12). Masses are shown as mean ± SEM. i, Whole-body cholesterol content from F/F control and L-A/C KO mice on chow diet at 15 weeks old (n = 5/7). All data are presented as mean ± SEM. P values were determined by two-sided Student’s t-test (b, d, g and i), two-sided Student’s t-test with Benjamini, Krieger and Yekutieli correction (c and e), or two-way ANOVA with Sidak’s correction for multiple comparisons (h).

Source data

Extended Data Fig. 5 Model for the role of hepatic Asters in liver and systemic cholesterol homeostasis.

In normal physiology (left side of schematic), fasting stimulates fatty acid release from adipose tissue to promote hepatic sphingomyelinase activity, which liberates sequestered cholesterol in the hepatocyte PM. Aster proteins recognize this newly accessible cholesterol and transport it to the ER for CE formation, suppression of SREBP-2 pathway targets, bile acid synthesis and VLDL production. Hepatic Asters are also induced by FXR and function in the RCT pathway by moving HDL-derived cholesterol (and LDL-derived cholesterol) within hepatocytes. Loss of hepatic Aster function (right side of the schematic) impairs CE formation and VLDL output during fasting. Loss of Asters in the liver also decreases the appearance of HDL-derived cholesterol in bile and feces, raises plasma cholesterol levels (due to enhanced liver cholesterol efflux), and causes peripheral cholesterol accumulation (for example, adrenal gland, brown adipose tissue).

Extended Data Fig. 6 Western blot quantifications.

a, Western blot quantifications of ALOD4 binding from Fig. 1c. Normalized to loading control Actin (n = 1). b, Western blot quantifications of indicated proteins from Fig. 1c. Normalized to sample processing control calnexin (n = 5). c, Western blot quantifications of indicated proteins from Fig. 2g. Normalized to sample processing controls calnexin or lamin a/c (n = 4). d, Western blot quantifications of ALOD4 binding from Fig. 3c. Normalized to loading control actin (n = 1). e and f, Western blot quantifications of SMPD3 from Fig. 4b and Fig. 4c. Normalized to loading control actin (n = 3). g, Western blot quantifications of indicated proteins from Fig. 4i. Equal amounts of protein was loaded for each line (n = 4). h, Western blot quantifications of indicated proteins from Fig. 5b. Normalized to sample processing control calnexin (n = 4). i, Western blot quantifications of indicated proteins from Fig. 5e. Normalized to actin which served as a loading control for SRB1 and a sample processing control for Aster-C (n = 1). j, Western blot quantifications of ABCA1 from Fig. 8c. Normalized to sample processing control calnexin (n = 5). k, l and m, Western blot quantifications of ALOD4 binding from extended data Figs. 3b, 3d and 3e. Normalized to loading control actin or calnexin (n = 1). n, Western blot quantifications of ALOD4 binding and Sr-B1 from Fig. 4a. Normalized to calnexin which served as a loading control for ALOD4 and a sample processing control for SRB1 (n = 1). Data are presented as mean ± SEM. P values were determined by two-sided Student’s t-test (b, c, e, f and h).

Source data

Supplementary information

Supplementary Information

Supplementary Table 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots/images/gating strategy.

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Fig. 7

Numerical source data.

Source Data Fig. 8

Unprocessed western blots/images.

Source Data Fig. 8

Numerical source data.

Source Data Extended Data Fig. 1

Numerical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots/gels.

Source Data Extended Data Fig. 2

Numerical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots/images.

Source Data Extended Data Fig. 3

Numerical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots/images.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots/images.

Source Data Extended Data Fig. 6

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

Xiao, X., Kennelly, J.P., Ferrari, A. et al. Hepatic nonvesicular cholesterol transport is critical for systemic lipid homeostasis. Nat Metab 5, 165–181 (2023). https://doi.org/10.1038/s42255-022-00722-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00722-6

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