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:

Liver macrophages regulate systemic metabolism through non-inflammatory factors

21 December 2022 Editor’s Note: Readers are alerted that the reliability of data presented in this article is currently in question. Appropriate editorial action will be taken once this matter is resolved.

An Author Correction to this article was published on 19 January 2021

An Author Correction to this article was published on 04 April 2019

This article has been updated

Abstract

Liver macrophages (LMs) have been proposed to contribute to metabolic disease through secretion of inflammatory cytokines. However, anti-inflammatory drugs lead to only modest improvements in systemic metabolism. Here we show that LMs do not undergo a proinflammatory phenotypic switch in obesity-induced insulin resistance in flies, mice and humans. Instead, we find that LMs produce non-inflammatory factors, such as insulin-like growth factor–binding protein 7 (IGFBP7), that directly regulate liver metabolism. IGFBP7 binds to the insulin receptor and induces lipogenesis and gluconeogenesis via activation of extracellular-signal-regulated kinase (ERK) signalling. We further show that IGFBP7 is subject to RNA editing at a higher frequency in insulin-resistant than in insulin-sensitive obese patients (90% versus 30%, respectively), resulting in an IGFBP7 isoform with potentially higher capacity to bind to the insulin receptor. Our study demonstrates that LMs can contribute to insulin resistance independently of their inflammatory status and indicates that non-inflammatory factors produced by macrophages might represent new drug targets for the treatment of metabolic diseases.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Obesity-induced insulin resistance occurs independently of inflammation.
Fig. 2: High-fat feeding increases Igfbp7 expression in LMs.
Fig. 3: Phenotype of Igfbp7-deficient LMs.
Fig. 4: Silencing of Igfbp7 in LMs decreases hyperglycaemia and hepatic steatosis.
Fig. 5: LM-derived IGFBP7 increases lipogenesis and gluconeogenesis.
Fig. 6: Proposed models for IGFBP7 signalling in mice and humans.

Similar content being viewed by others

Data availability

The data that support the plots within this paper and other findings of this study are available from the corresponding author upon reasonable request. Custom codes used for GSEA are available upon request. All sequence data for this study have been deposited in the Sequence Read Archive (SRA) under accession numbers PRJNA483744 (mouse data) and PRJNA491664 (human data).

Change history

  • 21 December 2022

    Editor’s Note: Readers are alerted that the reliability of data presented in this article is currently in question. Appropriate editorial action will be taken once this matter is resolved.

  • 04 April 2019

    In the version of this article initially published, author Volker M. Lauschke had affiliation number 13; the correct affiliation number is 12. The error has been corrected in the HTML and PDF versions of the article.

  • 19 January 2021

    A Correction to this paper has been published: https://doi.org/10.1038/s42255-021-00343-5

References

  1. Moore, M. C., Coate, K. C., Winnick, J. J., An, Z. & Cherrington, A. D. Regulation of hepatic glucose uptake and storage in vivo. Adv. Nutr.3, 286–294 (2012).

    Article  CAS  Google Scholar 

  2. Petersen, M. C., Vatner, D. F. & Shulman, G. I. Regulation of hepatic glucose metabolism in health and disease. Nat. Rev. Endocrinol.13, 572–587 (2017).

    Article  CAS  Google Scholar 

  3. Lackey, D. E. & Olefsky, J. M. Regulation of metabolism by the innate immune system. Nat. Rev. Endocrinol.12, 15–28 (2016).

    Article  CAS  Google Scholar 

  4. Tilg, H. & Moschen, A. R. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology52, 1836–1846 (2010).

    Article  CAS  Google Scholar 

  5. Jager, J., Aparicio-Vergara, M. & Aouadi, M. Liver innate immune cells and insulin resistance: the multiple facets of Kupffer cells. J. Intern. Med.280, 209–220 (2016).

    Article  CAS  Google Scholar 

  6. Esser, N., Paquot, N. & Scheen, A. J. Anti-inflammatory agents to treat or prevent type 2 diabetes, metabolic syndrome and cardiovascular disease. Expert Opin. Investig. Drugs24, 283–307 (2015).

    Article  CAS  Google Scholar 

  7. Pollack, R. M., Donath, M. Y., LeRoith, D. & Leibowitz, G. Anti-inflammatory agents in the treatment of diabetes and Its vascular complications. Diabetes Care39(Suppl. 2), S244–S252 (2016).

    Article  CAS  Google Scholar 

  8. Everett, B. M. et al. Anti-Inflammatory therapy with canakinumab for the prevention and management of diabetes. J. Am. Coll. Cardiol.71, 2392–2401 (2018).

    Article  CAS  Google Scholar 

  9. Chen, L. et al. Selective depletion of hepatic Kupffer cells significantly alleviated hepatosteatosis and intrahepatic inflammation induced by high fat diet. Hepatogastroenterology59, 1208–1212 (2012).

    CAS  Google Scholar 

  10. Clementi, A. H., Gaudy, A. M., van Rooijen, N., Pierce, R. H. & Mooney, R. A. Loss of Kupffer cells in diet-induced obesity is associated with increased hepatic steatosis, STAT3 signaling, and further decreases in insulin signaling. Biochim. Biophys. Acta1792, 1062–1072 (2009).

    Article  CAS  Google Scholar 

  11. Lanthier, N. et al. Kupffer cell activation is a causal factor for hepatic insulin resistance. Am. J. Physiol. Gastrointest. Liver Physiol.298, G107–G116 (2010).

    Article  CAS  Google Scholar 

  12. Papackova, Z. et al. Kupffer cells ameliorate hepatic insulin resistance induced by high-fat diet rich in monounsaturated fatty acids: the evidence for the involvement of alternatively activated macrophages. Nutr. Metab.9, 22 (2012).

    Article  CAS  Google Scholar 

  13. Stienstra, R. et al. Kupffer cells promote hepatic steatosis via interleukin-1β-dependent suppression of peroxisome proliferator-activated receptor α activity. Hepatology51, 511–522 (2010).

    Article  CAS  Google Scholar 

  14. Figueroa-Clarevega, A. & Bilder, D. Malignant Drosophila tumors interrupt insulin signaling to induce cachexia-like wasting. Dev. Cell33, 47–55 (2015).

    Article  CAS  Google Scholar 

  15. Kwon, Y. et al. Systemic organ wasting induced by localized expression of the secreted insulin/IGF antagonist ImpL2. Dev. Cell33, 36–46 (2015).

    Article  CAS  Google Scholar 

  16. Evdokimova, V. et al. IGFBP7 binds to the IGF-1 receptor and blocks its activation by insulin-like growth factors. Sci. Signal.5, ra92 (2012).

    Article  Google Scholar 

  17. Kratz, M. et al. Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab.20, 614–625 (2014).

    Article  CAS  Google Scholar 

  18. Hardy, O. T. et al. Body mass index–independent inflammation in omental adipose tissue associated with insulin resistance in morbid obesity. Surg. Obes. Relat. Dis.7, 60–67 (2011).

    Article  Google Scholar 

  19. Haukeland, J. W. et al. Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. J. Hepatol.44, 1167–1174 (2006).

    Article  CAS  Google Scholar 

  20. Ahrens, M. et al. DNA methylation analysis in nonalcoholic fatty liver disease suggests distinct disease-specific and remodeling signatures after bariatric surgery. Cell Metab.18, 296–302 (2013).

    Article  CAS  Google Scholar 

  21. Horvath, S. et al. Obesity accelerates epigenetic aging of human liver. Proc. Natl Acad. Sci. USA111, 15538–15543 (2014).

    Article  CAS  Google Scholar 

  22. Morinaga, H. et al. Characterization of distinct subpopulations of hepatic macrophages in HFD/obese mice. Diabetes64, 1120–1130 (2015).

    Article  CAS  Google Scholar 

  23. Aouadi, M. et al. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature458, 1180–1184 (2009).

    Article  CAS  Google Scholar 

  24. Tesz, G. J. et al. Glucan particles for selective delivery of siRNA to phagocytic cells in mice. Biochem. J.436, 351–362 (2011).

    Article  CAS  Google Scholar 

  25. Tencerova, M. et al. Activated Kupffer cells inhibit insulin sensitivity in obese mice. FASEB J.29, 2959–2969 (2015).

    Article  CAS  Google Scholar 

  26. Wan, F. & Lenardo, M. J. The nuclear signaling of NF-κB: current knowledge, new insights, and future perspectives. Cell Res.20, 24–33 (2010).

    Article  CAS  Google Scholar 

  27. Fang, B. et al. Circadian enhancers coordinate multiple phases of rhythmic gene transcription in vivo. Cell159, 1140–1152 (2014).

    Article  CAS  Google Scholar 

  28. Aparicio-Vergara, M., Tencerova, M., Morgantini, C., Barreby, E. & Aouadi, M. Isolation of Kupffer cells and hepatocytes from a single mouse liver. Methods Mol. Biol.1639, 161–171 (2017).

    Article  CAS  Google Scholar 

  29. Huang, W. et al. Depletion of liver Kupffer cells prevents the development of diet-induced hepatic steatosis and insulin resistance. Diabetes59, 347–357 (2010).

    Article  CAS  Google Scholar 

  30. Halpern, K. B. et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature542, 352–356 (2017).

    Article  CAS  Google Scholar 

  31. Scott, C. L. et al. Bone marrow–derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat. Commun.7, 10321 (2016).

    Article  CAS  Google Scholar 

  32. Levanon, E. Y. et al. Evolutionarily conserved human targets of adenosine to inosine RNA editing. Nucleic Acids Res.33, 1162–1168 (2005).

    Article  CAS  Google Scholar 

  33. Godfried Sie, C., Hesler, S., Maas, S. & Kuchka, M. IGFBP7’s susceptibility to proteolysis is altered by A-to-I RNA editing of its transcript. FEBS Lett.586, 2313–2317 (2012).

    Article  CAS  Google Scholar 

  34. Ahmed, S. et al. Proteolytic processing of IGFBP-related protein-1 (TAF/angiomodulin/mac25) modulates its biological activity. Biochem. Biophys. Res. Commun.310, 612–618 (2003).

    Article  CAS  Google Scholar 

  35. Bader, R. et al. The IGFBP7 homolog Imp-L2 promotes insulin signaling in distinct neurons of the Drosophila brain. J. Cell Sci.126, 2571–2576 (2013).

    CAS  Google Scholar 

  36. Jiao, P., Feng, B., Li, Y., He, Q. & Xu, H. Hepatic ERK activity plays a role in energy metabolism. Mol. Cell Endocrinol.375, 157–166 (2013).

    Article  CAS  Google Scholar 

  37. Han, M. S. et al. JNK expression by macrophages promotes obesity-induced insulin resistance and inflammation. Science339, 218–222 (2013).

    Article  CAS  Google Scholar 

  38. Cai, D. et al. Local and systemic insulin resistance resulting from hepatic activation of IKK-β and NF-κB. Nat. Med.11, 183–190 (2005).

    Article  CAS  Google Scholar 

  39. Obstfeld, A. E. et al. C-C chemokine receptor 2 (CCR2) regulates the hepatic recruitment of myeloid cells that promote obesity-induced hepatic steatosis. Diabetes59, 916–925 (2010).

    Article  CAS  Google Scholar 

  40. Arkan, M. C. et al. IKK-β links inflammation to obesity-induced insulin resistance. Nat. Med.11, 191–198 (2005).

    Article  CAS  Google Scholar 

  41. Odegaard, J. I. et al. Alternative M2 activation of Kupffer cells by PPARδ ameliorates obesity-induced insulin resistance. Cell Metab.7, 496–507 (2008).

    Article  CAS  Google Scholar 

  42. Lanthier, N. et al. Kupffer cell depletion prevents but has no therapeutic effect on metabolic and inflammatory changes induced by a high-fat diet. FASEB J.25, 4301–4311 (2011).

    Article  CAS  Google Scholar 

  43. Honegger, B. et al. Imp-L2, a putative homolog of vertebrate IGF-binding protein 7, counteracts insulin signaling in Drosophila and is essential for starvation resistance. J. Biol.7, 10 (2008).

    Article  Google Scholar 

  44. Ussar, S., Bezy, O., Bluher, M. & Kahn, C. R. Glypican-4 enhances insulin signaling via interaction with the insulin receptor and serves as a novel adipokine. Diabetes61, 2289–2298 (2012).

    Article  CAS  Google Scholar 

  45. Boothe, T. et al. Inter-domain tagging implicates caveolin-1 in insulin receptor trafficking and Erk signaling bias in pancreatic beta-cells. Mol. Metab.5, 366–378 (2016).

    Article  CAS  Google Scholar 

  46. Belfiore, A., Frasca, F., Pandini, G., Sciacca, L. & Vigneri, R. Insulin receptor isoforms and insulin receptor/insulin-like growth factor receptor hybrids in physiology and disease. Endocr. Rev.30, 586–623 (2009).

    Article  CAS  Google Scholar 

  47. Leibiger, B. et al. PI3K-C2α knockdown results in rerouting of insulin signaling and pancreatic beta cell proliferation. Cell Rep.13, 15–22 (2015).

    Article  CAS  Google Scholar 

  48. Kumar, S., Bradley, C. L., Mukashyaka, P. & Doerrler, W. T. Identification of essential arginine residues of Escherichiacoli DedA/Tvp38 family membrane proteins YqjA and YghB. FEMS Microbiol. Lett.363, fnw133 (2016).

  49. Dahms, S. O., Hardes, K., Steinmetzer, T. & Than, M. E. X-ray structures of the proprotein convertase furin bound with substrate analogue inhibitors reveal substrate specificity determinants beyond the S4 pocket. Biochemistry57, 925–934 (2018).

    Article  CAS  Google Scholar 

  50. Eisenberg, E. & Levanon, E. Y. A-to-I RNA editing—immune protector and transcriptome diversifier. Nat. Rev. Genet.19, 473–490 (2018).

    Article  CAS  Google Scholar 

  51. Donath, M. Y. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat. Rev. Drug Discov.13, 465–476 (2014).

    Article  CAS  Google Scholar 

  52. Lee, J. H. et al. Hepatic steatosis index: a simple screening tool reflecting nonalcoholic fatty liver disease. Dig. Liver Dis.42, 503–508 (2010).

    Article  CAS  Google Scholar 

  53. Bell, C. C. et al. Transcriptional, functional, and mechanistic comparisons of stem cell–derived hepatocytes, HepaRG cells, and three-dimensional human hepatocyte spheroids as predictive in vitro systems for drug-induced liver injury. Drug Metab. Dispos.45, 419–429 (2017).

    Article  CAS  Google Scholar 

  54. Wijkstrom, J. et al. Renal morphology, clinical findings, and progression rate in Mesoamerican nephropathy. Am. J. Kidney Dis.69, 626–636 (2017).

    Article  Google Scholar 

  55. Weibel, E. R. Stereological methods in cell biology: where are we—where are we going? J. Histochem. Cytochem.29, 1043–1052 (1981).

    Article  CAS  Google Scholar 

  56. Parini, P., Johansson, L., Broijersen, A., Angelin, B. & Rudling, M. Lipoprotein profiles in plasma and interstitial fluid analyzed with an automated gel-filtration system. Eur. J. Clin. Invest.36, 98–104 (2006).

    Article  CAS  Google Scholar 

  57. Tennessen, J. M., Barry, W. E., Cox, J. & Thummel, C. S. Methods for studying metabolism in Drosophila. Methods68, 105–115 (2014).

    Article  CAS  Google Scholar 

  58. Schneedorferova, I., Tomcala, A. & Valterova, I. Effect of heat treatment on the n-3/n-6 ratio and content of polyunsaturated fatty acids in fish tissues. Food Chem.176, 205–211 (2015).

    Article  CAS  Google Scholar 

  59. Kutsukake, M. et al. Circulating IGF-binding protein 7 (IGFBP7) levels are elevated in patients with endometriosis or undergoing diabetic hemodialysis. Reprod. Biol. Endocrinol.6, 54 (2008).

    Article  Google Scholar 

  60. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol.14, R36 (2013).

    Article  Google Scholar 

  61. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics30, 923–930 (2014).

    Article  CAS  Google Scholar 

  62. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics26, 841–842 (2010).

    Article  CAS  Google Scholar 

  63. 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  Google Scholar 

  64. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol.11, R25 (2010).

    Article  Google Scholar 

  65. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc.57, 289–300 (1995).

    Google Scholar 

  66. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet.34, 267 (2003).

    Article  CAS  Google Scholar 

  67. Falcon, S. & Gentleman, R. Using GOstats to test gene lists for GO term association. Bioinformatics23, 257–258 (2007).

    Article  CAS  Google Scholar 

  68. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets—update. Nucleic Acids Res.41, D991–D995 (2013).

    Article  CAS  Google Scholar 

  69. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res.43, e47 (2015).

    Article  Google Scholar 

  70. Rosner, B. Fundamental of Biostatistics 7th edn (Brooks/Cole CENGAGE Learning, 2010).

Download references

Acknowledgements

We are grateful to R. Harris, S. Craige and B. Craige for their considerable input on the manuscript. We thank T. Dolezal and M. Jindra for helpful suggestions on the Drosophila work. We also thank T. Schröder and R. Kuiper for the tissue histology, M. Taipale and J. Liu for performing sequencing, and A. Krstic for providing flow cytometry support. We thank M. Nordstrand for organising the collection of human obese liver biopsies. This work was supported by research grants from AstraZeneca through the ICMC (M.A.), the Swedish Research Council (M.A.; 2015-03582), the Stockholm County Council (E.N.), the Novo Nordisk Foundation, including the Tripartite Immuno-metabolism Consortium (M.A. and TrIC; NNF15CC0018486), the Strategic Research Program in Diabetes at Karolinska Institutet (M.A. and E.N.), the Diabetes Wellness Foundation Sweden (J.J.) and the Ruth and Richard Julin Foundation (C.M.).

Author information

Authors and Affiliations

Authors

Contributions

C.M. carried out most experiments. G.K. and A.B. performed and analysed the data from the Drosophila experiments. J.J., L.L., V.A., E.B., A.S., C.X., M.T., C.K. and F.V. helped with experiments and interpretation of the data. S.S. measured circulating lipid levels in mice. A.T. measured lipids in Drosophila. K.H. generated all electron microscopy images and measurements. N.K.B. and E.E. provided biopsies and liver cells from lean individuals. E.N. provided liver biopsies from obese human subjects. M.R. provided serum from obese and lean volunteers. T.H. and V.M.L. performed experiments in human liver spheroids. X.L. and C.K. performed the bioinformatics analysis and interpretation. J.B. contributed to the design of experiments related to IGFBP7 signalling. C.M. and M.A. conceived the project, analysed data and wrote the manuscript with input from all co-authors.

Corresponding author

Correspondence to Myriam Aouadi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figures 1–8 and Supplementary Tables 1–8

Reporting Summary

Supplementary Data 1

Significantly regulated pathways in human LMs isolated from obese as compared to lean patients. GSEA depicting significantly up- or downregulated pathways (FDR < 0.05) in human LMs according to MES.

Supplementary Data 2

Significantly regulated pathways in LMs isolated from HFD-fed mice as compared to ND-fed mice. GSEA depicting significantly up- or downregulated pathways (FDR < 0.05) in mouse LMs according to MES.

Supplementary Data 3

Obesity-regulated genes in mouse LMs and in mouse whole liver. List of regulated genes in LMs and in whole liver from mice fed a HFD as compared to mice fed a ND. Data include gene name, log2 fold change (log2FC), P value and FDR.

Supplementary Data 4

Highly expressed and upregulated genes in LMs isolated from HFD-fed mice as compared to ND-fed mice. List of upregulated genes in LMs isolated from HFD-fed as compared to ND-fed mice with median RPKM > 50. Data include gene name, log2 fold change (log2FC), P value and FDR.

Supplementary Data 5

Gene expression regulated by obesity and insulin resistance in human LMs. List of regulated genes in LMs isolated from humans: obese as compared to lean patients and obese insulin-resistant as compared to obese insulin-sensitive patients. Data include gene name, log2 fold change (log2FC), P value and FDR.

Supplementary Data 6

Gene expression regulated in mouse LMs following Igfbp7 silencing. List of regulated genes in LMs isolated from mice treated with GeRP-Scr versus GeRP-Igfbp7. Data includes gene name, log2 fold change (log2FC), P value and FDR.

Supplementary Data 7

Gene ontology enrichment analysis of inversely regulated genes in LMs from HFD- versus ND-fed mice and mice treated with GeRP-Igfbp7 versus GeRP-Scr. Complete list of inversely regulated pathways in mice fed a HFD versus ND as compared to mice treated with GeRP-Igfbp7 versus GeRP-Scr.

Supplementary Data 8

Genes regulated in mouse hepatocytes following Igfbp7 silencing in LMs. List of regulated genes in hepatocytes isolated from mice treated with GeRP-Scr versus GeRP-Igfbp7. Data include gene name, log2 fold change (log2FC), P value and FDR.

Supplementary Data 9

Significantly regulated pathways in mouse hepatocytes following Igfbp7 silencing in LMs. GSEA depicting significantly up- or downregulated pathways (FDR < 0.05) in human and mouse LMs according to MES.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morgantini, C., Jager, J., Li, X. et al. Liver macrophages regulate systemic metabolism through non-inflammatory factors. Nat Metab 1, 445–459 (2019). https://doi.org/10.1038/s42255-019-0044-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-019-0044-9

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