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.

  • Letter
  • Published:

Dicer1–miR-328–Bace1 signalling controls brown adipose tissue differentiation and function

Abstract

Activation of brown adipose tissue (BAT) controls energy homeostasis in rodents and humans and has emerged as an innovative strategy for the treatment of obesity and type 2 diabetes mellitus1,2,3,4. Here we show that ageing- and obesity-associated dysfunction of brown fat coincides with global microRNA downregulation due to reduced expression of the microRNA-processing node Dicer1. Consequently, heterozygosity of Dicer1 in BAT aggravated diet-induced-obesity (DIO)-evoked deterioration of glucose metabolism. Analyses of differential microRNA expression during preadipocyte commitment and mouse models of progeria, longevity and DIO identified miR-328 as a regulator of BAT differentiation. Reducing miR-328 blocked preadipocyte commitment, whereas miR-328 overexpression instigated BAT differentiation and impaired muscle progenitor commitment—partly through silencing of the β-secretase Bace1. Loss of Bace1 enhanced brown preadipocyte specification in vitro and was overexpressed in BAT of obese and progeroid mice. In vivo Bace1 inhibition delayed DIO-induced weight gain and improved glucose tolerance and insulin sensitivity. These experiments reveal Dicer1–miR-328–Bace1 signalling as a determinant of BAT function, and highlight the potential of Bace1 inhibition as a therapeutic approach to improve not only neurodegenerative diseases but also ageing- and obesity-associated impairments of BAT function.

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

Figure 1: Brown-fat-specific Dicer1 haploinsufficiency recapitulates obesity- and ageing-associated decline in microRNA expression.
Figure 2: Partial Dicer1 deficiency in BAT predisposes mice to obesity-associated deterioration of glucose homeostasis.
Figure 3: In vitro/in vivo filtering approach identifies miR-328 as a BAT-regulatory microRNA.
Figure 4: miR-328 and miR-193b support brown adipogenesis and repress myoblast differentiation in vitro, partly through silencing of Bace1.
Figure 5: In vivo inhibition of Bace1 activates BAT and ameliorates obesity-associated deterioration of glucose metabolism.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Bartelt, A. & Heeren, J. Adipose tissue browning and metabolic health. Nat. Rev. Endocrinol. 10, 24–36 (2014).

    Article  CAS  Google Scholar 

  2. Virtanen, K. A. et al. Functional brown adipose tissue in healthy adults. N. Engl. J. Med. 360, 1518–1525 (2009).

    Article  CAS  Google Scholar 

  3. van Marken Lichtenbelt, W. D. et al. Cold-activated brown adipose tissue in healthy men. N. Engl. J. Med. 360, 1500–1508 (2009).

    Article  CAS  Google Scholar 

  4. Yoneshiro, T. et al. Recruited brown adipose tissue as an antiobesity agent in humans. J. Clin. Invest. 123, 3404–3408 (2013).

    Article  CAS  Google Scholar 

  5. Kennedy, G. C. The role of depot fat in the hypothalamic control of food intake in the rat. Proc. R. Soc. Lond. B 140, 578–596 (1953).

    Article  CAS  Google Scholar 

  6. Cannon, B. & Nedergaard, J. Thermogenesis challenges the adipostat hypothesis for body-weight control. Proc. Nutr. Soc. 68, 401–407 (2009).

    Article  Google Scholar 

  7. Lowell, B. B. et al. Development of obesity in transgenic mice after genetic ablation of brown adipose tissue. Nature 366, 740–742 (1993).

    Article  CAS  Google Scholar 

  8. Kajimura, S. et al. Initiation of myoblast to brown fat switch by a PRDM16-C/EBP-β transcriptional complex. Nature 460, 1154–1158 (2009).

    Article  CAS  Google Scholar 

  9. Kajimura, S., Seale, P. & Spiegelman, B. M. Transcriptional control of brown fat development. Cell Metab. 11, 257–262 (2010).

    Article  CAS  Google Scholar 

  10. Carthew, R. W. Gene regulation by microRNAs. Curr. Opin. Genet. Dev. 16, 203–208 (2006).

    Article  CAS  Google Scholar 

  11. Chen, Y. et al. miR-155 regulates differentiation of brown and beige adipocytes via a bistable circuit. Nat. Commun. 4, 1769 (2013).

    Article  Google Scholar 

  12. Sun, L. et al. Mir193b-365 is essential for brown fat differentiation. Nat. Cell Biol. 13, 958–965 (2012).

    Article  Google Scholar 

  13. Trajkovski, M., Ahmed, K., Esau, C. C. & Stoffel, M. MyomiR-133 regulates brown fat differentiation through Prdm16. Nat. Cell Biol. 14, 1330–1335 (2012).

    Article  CAS  Google Scholar 

  14. Mattson, M. P. Perspective: does brown fat protect against diseases of aging? Ageing Res. Rev. 9, 69–76 (2010).

    Article  CAS  Google Scholar 

  15. Rogers, N. H., Landa, A., Park, S. & Smith, R. G. Aging leads to a programmed loss of brown adipocytes in murine subcutaneous white adipose tissue. Aging Cell 11, 1074–1083 (2012).

    Article  CAS  Google Scholar 

  16. Mori, M. A. et al. Altered miRNA processing disrupts brown/white adipocyte determination and associates with lipodystrophy. J. Clin. Invest. 124, 3339–3351 (2014).

    Article  CAS  Google Scholar 

  17. Mori, M. A. et al. Role of microRNA processing in adipose tissue in stress defense and longevity. Cell Metab. 16, 336–347 (2012).

    Article  CAS  Google Scholar 

  18. Shimizu, I. et al. Vascular rarefaction mediates whitening of brown fat in obesity. J. Clin. Invest. 124, 2099–2112 (2014).

    Article  CAS  Google Scholar 

  19. Rupaimoole, R. et al. Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression. Nat. Commun. 5, 5202 (2014).

    Article  CAS  Google Scholar 

  20. van den Beucken, T. et al. Hypoxia promotes stem cell phenotypes and poor prognosis through epigenetic regulation of DICER. Nat. Commun. 5, 5203 (2014).

    Article  CAS  Google Scholar 

  21. Mudhasani, R. et al. Dicer is required for the formation of white but not brown adipose tissue. J. Cell Physiol. 226, 1399–1406 (2011).

    Article  CAS  Google Scholar 

  22. Turpin, S. M. et al. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab. 20, 678–686 (2014).

    Article  CAS  Google Scholar 

  23. Schulz, T. J. et al. Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature 495, 379–383 (2013).

    Article  CAS  Google Scholar 

  24. Guerra, C. et al. Brown adipose tissue-specific insulin receptor knockout shows diabetic phenotype without insulin resistance. J. Clin. Invest. 108, 1205–1213 (2001).

    Article  CAS  Google Scholar 

  25. Bartelt, A. et al. Brown adipose tissue activity controls triglyceride clearance. Nat. Med. 17, 200–205 (2011).

    Article  CAS  Google Scholar 

  26. Feldmann, H. M., Golozoubova, V., Cannon, B. & Nedergaard, J. UCP1 ablation induces obesity and abolishes diet-induced thermogenesis in mice exempt from thermal stress by living at thermoneutrality. Cell Metab. 9, 203–209 (2009).

    Article  CAS  Google Scholar 

  27. Kopecky, J., Hodny, Z., Rossmeisl, M., Syrovy, I. & Kozak, L. P. Reduction of dietary obesity in aP2-Ucp transgenic mice: physiology and adipose tissue distribution. Am. J. Physiol. 270, E768–E775 (1996).

    CAS  PubMed  Google Scholar 

  28. Ohno, H., Shinoda, K., Ohyama, K., Sharp, L. Z. & Kajimura, S. EHMT1 controls brown adipose cell fate and thermogenesis through the PRDM16 complex. Nature 504, 163–167 (2013).

    Article  CAS  Google Scholar 

  29. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    Article  CAS  Google Scholar 

  30. Ho, L. et al. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J. 18, 902–904 (2004).

    Article  CAS  Google Scholar 

  31. Boissonneault, V., Plante, I., Rivest, S. & Provost, P. MicroRNA-298 and microRNA-328 regulate expression of mouse β-amyloid precursor protein-converting enzyme 1. J. Biol. Chem. 284, 1971–1981 (2009).

    Article  CAS  Google Scholar 

  32. Meakin, P. J. et al. Reduction in BACE1 decreases body weight, protects against diet-induced obesity and enhances insulin sensitivity in mice. Biochem. J. 441, 285–296 (2012).

    Article  CAS  Google Scholar 

  33. Hilpert, H. et al. β-Secretase (BACE1) inhibitors with high in vivo efficacy suitable for clinical evaluation in Alzheimer’s disease. J. Med. Chem. 56, 3980–3995 (2013).

    Article  CAS  Google Scholar 

  34. Vignini, A. et al. Alzheimer’s disease and diabetes: new insights and unifying therapies. Curr. Diab. Rev. 9, 218–227 (2013).

    Article  Google Scholar 

  35. Belgardt, B. F. et al. Hypothalamic and pituitary c-Jun N-terminal kinase 1 signaling coordinately regulates glucose metabolism. Proc. Natl Acad. Sci. USA 107, 6028–6033 (2010).

    Article  CAS  Google Scholar 

  36. Kornfeld, J. W. et al. Obesity-induced overexpression of miR-802 impairs glucose metabolism through silencing of Hnf1b. Nature 494, 111–115 (2013).

    Article  CAS  Google Scholar 

  37. Gao, J. et al. CLUH regulates mitochondrial biogenesis by binding mRNAs of nuclear-encoded mitochondrial proteins. J. Cell Biol. 207, 213–223 (2014).

    Article  CAS  Google Scholar 

  38. Engblom, D. et al. Direct glucocorticoid receptor-Stat5 interaction in hepatocytes controls body size and maturation-related gene expression. Genes Dev. 21, 1157–1162 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Hampel, A. Schauss, H. Jacobsen, J. Altmüller and J. Alber for technical assistance and A. Tarakhovsky (Rockefeller University, USA) for providing Dicer1loxP/loxP mice. We are grateful for Amesdf mice from A. Bartke (Southern Illinois University School of Medicine, USA). J.-W.K., M.O., N.H. and S.Konieczka are supported by the Emmy-Noether Program (DFG; KO4728/1.1) and CECAD. E.S. is supported by Evangelisches Studienwerk Villigst. S.Khani appreciates support from DAAD. E.T. and M.A.M. are supported by CNPq, CAPES and FAPESP (2010/52557-0). J.C.B. is supported by the Leibniz Preis (BR1492/7-1), CECAD, Cologne Center for Molecular Medicine Cologne (CMMC) and through financial support from ‘Systems Biology of Ageing Cologne’ (Sybacol).

Author information

Authors and Affiliations

Authors

Contributions

M.O., E.S., J.M., C.B., N.H., S.Khani, S.Konieczka, M.P.-J., S.B., T.-M.V., D.B., H.S.B., M.H., E.T., P.F. and J.-W.K. performed experiments. H.H., M.A.M., J.C.B. and J.-W.K. designed the experimental set-up. H.H., J.C.B., G.A.G. and J.H. contributed discussions.

Corresponding author

Correspondence to Jan-Wilhelm Kornfeld.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1

(a) Relative expression of Cidea, Dio2, Elovl3 and Ucp1 in BAT of adult (6 months, n = 4) versus old (18 months, n = 5) mice (left). Photomicrograph of BAT from adult versus old mice. A representative image is shown (right). (b) Network illustration of IPA pathways changes on RNA-Seq based mRNA expression in BAT in NCD-fed (n = 4) versus HFD-fed (n = 4) male C57BL/6 mice. (c) Breeding scheme for the generation of BAT-specific Dicer1 heterozygous knockouts (Dicer1ΔBAT/+). (d) Relative Ucp1 expression in SCAT of Dicer1ΔBAT/+ mice (n = 5) versus control mice (n = 3). If not stated otherwise, boxplots indicate median ± min/max and an unpaired two-tailed Student’s t-test was used to assess statistical significance. *p < 0.05.

Supplementary Figure 2

(a) Body weight curves of female, NCD-fed Dicer1ΔBAT/+ (n = 11; black circle) versus control (n = 11; white circle) or female, HFD-fed Dicer1ΔBAT/+ (n = 10, black diamond) versus control (n = 13; white diamond) mice. No statistically significant interaction was found using two-way ANOVAs for repeated measures (2WA-RM). (b) Relative expression of indicated genes in BAT of male, NCD-fed Dicer1ΔBAT/+ mice (n = 7) versus control mice (n = 8).(c) Core body temperature on cold (4 °C) exposure for indicated time points of female, Dicer1ΔBAT/+ (n = 7) versus control (n = 13) mice determined by rectal thermometry. No statistically significant interaction was found using 2WA-RM. (d) Glucose tolerance test of NCD-fed, male Dicer1ΔBAT/+ (n = 13) versus control animals (n = 20, left) or NCD-fed, female Dicer1ΔBAT/+ (n = 14) versus control animals (n = 21, right). A statistically significant interaction was found using 2WA-RM for male (P < 0.01; F = 4.165; Df = 4) mice and a borderline significant interaction for female (P < 0.07; F = 2.222; Df = 4) mice. (e) Insulin tolerance test of NCD-fed, male Dicer1ΔBAT/+ (n = 13) versus control animals (n = 15, left) or NCD-fed, female Dicer1ΔBAT/+ (n = 14) versus control animals (n = 16, right). No statistically significant interaction was found using 2WA-RM. (f) Serum insulin level of NCD-fed, male Dicer1ΔBAT/+ (n = 9) versus control mice (n = 10) and HFD-fed, male Dicer1ΔBAT/+ (n = 5) versus control mice (n = 8, left) or NCD-fed, female Dicer1ΔBAT/+ (n = 8) versus control mice (n = 9) and HFD-fed, female Dicer1ΔBAT/+ (n = 5) versus control mice (n = 12, right). (g) Serum leptin level of NCD-fed, male Dicer1ΔBAT/+ (n = 10) versus control mice (n = 9) and HFD-fed, male Dicer1ΔBAT/+ (n = 7) versus control mice (n = 13, left) or NCD-fed, female Dicer1ΔBAT/+ (n = 9) versus control mice (n = 9) and HFD-fed, female Dicer1ΔBAT/+(n = 10) versus control mice (n = 11, right). (h) Tissue weight/body weight (BW) ratio of indicated tissues in HFD-fed Dicer1ΔBAT/+ (Liver, n = 10; BAT, n = 4; SCAT, n = 3; VAT, n = 11) versus control (Liver, n = 18; BAT, n = 4; SCAT, n = 4; VAT, n = 17) mice. (i) Thin-layer chromatography-mediated quantification of lipid species (Chol, cholesterole; FFA, free fatty acids; TG, triglycerides) in indicated tissues of HFD-fed Dicer1ΔBAT/+ (n = 5) versus control (n = 6) mice. (j) Photomicrograph of indicated tissues in HFD-fed Dicer1ΔBAT/+ and control mice. A representative image is shown. Scale bar, 100 μm. If not stated otherwise, dots indicate mean ± s.e.m., boxplots indicate median ± min/max and an unpaired two-tailed Student’s t-test was used to assess statistical significance. *p < 0.05, **p < 0.01, ***p < 0.001, p < 0.0001.

Supplementary Figure 3

(a) Expression of indicated genes in differentiated PIBA cells after transfection with LNAs against indicated microRNAs versus scramble (scr) controls across n = 3 (miR-96) or 2 independent experiments. Bars indicate the mean of the experiments. (b) Immunoblots against indicated proteins after transfection of PIBA cells with LNAs against indicated microRNAs versus scr. Image is representative of three (UCP1) or two (others) independent experiments. (c) Relative expression of indicated genes in 3T3-L1 cells six days after transfection with LNAs against indicated microRNAs versus scr controls or microRNA mimics versus scr. All experiments were performed in triplicate and represent the average of n = 3 independent experiments. (d) Immunoblots against indicated proteins after transfection of 3T3-L1 cells with microRNA LNAs versus scr or microRNA mimics versus scr. Image is representative of two independent experiments. If not stated otherwise, boxplots indicate median ± min/max and a paired two-tailed Student’s t-test was used to assess statistical significance. Uncropped images of blots are shown in Supplementary Fig. 6. Source data values for panel a are available in Supplementary Table 5.

Supplementary Figure 4

(a) Sequences of mmu-miR-193b and mmu-miR-328, comparison of miR-193b versus miR-328 seed sequences (top) and overlap of in silico predicted miR-193b (grey) and miR-328 mRNA (black) targets (bottom). The statistical significance for the overlap of miR-193b and miR-328 target genesets is given in Methods. (b) Relative luciferase activity in HEK293 cells after co-transfection of pmiRGlo reporter constructs harbouring wild-type 3′UTR fragments of indicated genes together with 100 nM of scr, miR-193b or miR-328 mimics across n = 3 independent experiments. (c) Relative expression of Bace1 (n = 6) and Gprc5b (n = 8) in indicated tissues (BAT, brown adipose tissue; H, heart; Liv, liver; SM, skeletal muscle; WAT, white adipose tissue). Expression levels in BAT were set to unity = 1. (d) Relative expression of Bace1 and Gprc5b in C2C12 cells seven days after transfection with 80 nM of indicated longRNA GapmeRs across n = 4 (Bace1) or n = 5 (Gprc5b) independent experiments, left). Relative expression of Bace1 in PIBA cells two days after transfection with 80 nM of indicated longRNA GapmeRs across n = 3 independent experiments, right). (e) Photomicrograph of C2C12 cells seven days after transfection with 80 nM of indicated longRNA Gapmers. Image is representative of three independent experiments. Scale bar, 100 μm. (f) Relative expression of Myf5, Myod1, Pax3 and Runx1t1 in C2C12 cells seven days after transfection with 80 nM of indicated longRNA GapmeRs. (g) Relative expression of indicated genes in 3T3-L1 cells two days after transfection with LNAs against Bace1 versus scr across n = 4 independent experiments. If not stated otherwise, boxplots indicate median ± min/max and a paired two-tailed Student’s t-test was used to assess statistical significance. * = p < 0.05.

Supplementary Figure 5

(a) Average food intake quantified by manual recordings of HFD-fed (n = 15) and HFD + RO5508887-fed (n = 15) mice during the course of the feeding experiment (12 weeks). (b) Glucose-stimulated insulin secretion of HFD-fed (n = 7) versus HFD + RO5508887-fed (n = 7) mice. (c) Percentage of insulin-positive (ins +) cells in pancreatic sections from HFD-fed (n = 6) and HFD + RO5508887-fed (n = 6) mice (left). Insulin staining and H&E counterstaining of pancreatic islets in HFD and HFD + RO5508887-fed mice (right). A representative image is shown. Scale bar, 100 μm. (d) Body composition measured by nuclear magnetic resonance of HFD-fed control (n = 15) versus HFD + RO5508887-fed (n = 14) male C57BL/6 mice. (e) H&E staining of SCAT paraffine section in HFD-fed (n = 5) and HFD + RO5508887-fed (n = 5) mice analyzed in using Zeiss Axiovision 4.2 software and adipocyte area cumulatively plotted in 10 increments. If not stated otherwise, boxplots indicate median ± min/max and an unpaired two-tailed Student’s t-test was used to assess statistical significance. *p < 0.05.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1740 kb)

Supplementary Table 1

Supplementary Information (XLSX 32 kb)

Supplementary Table 2

Supplementary Information (XLSX 11 kb)

Supplementary Table 3

Supplementary Information (XLSX 9 kb)

Supplementary Table 4

Supplementary Information (XLSX 10 kb)

Supplementary Table 5

Supplementary Information (XLSX 31 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Oliverio, M., Schmidt, E., Mauer, J. et al. Dicer1–miR-328–Bace1 signalling controls brown adipose tissue differentiation and function. Nat Cell Biol 18, 328–336 (2016). https://doi.org/10.1038/ncb3316

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3316

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