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:

SWELL1 is a regulator of adipocyte size, insulin signalling and glucose homeostasis

An Erratum to this article was published on 29 June 2017

An Erratum to this article was published on 31 May 2017

This article has been updated

Abstract

Adipocytes undergo considerable volumetric expansion in the setting of obesity. It has been proposed that such marked increases in adipocyte size may be sensed via adipocyte-autonomous mechanisms to mediate size-dependent intracellular signalling. Here, we show that SWELL1 (LRRC8a), a member of the Leucine-Rich Repeat Containing protein family, is an essential component of a volume-sensitive ion channel (VRAC) in adipocytes. We find that SWELL1-mediated VRAC is augmented in hypertrophic murine and human adipocytes in the setting of obesity. SWELL1 regulates adipocyte insulin–PI3K–AKT2–GLUT4 signalling, glucose uptake and lipid content via SWELL1 C-terminal leucine-rich repeat domain interactions with GRB2/Cav1. Silencing GRB2 in SWELL1 KO adipocytes rescues insulin-pAKT2 signalling. In vivo, shRNA-mediated SWELL1 knockdown and adipose-targeted SWELL1 knockout reduce adiposity and adipocyte size in obese mice while impairing systemic glycaemia and insulin sensitivity. These studies identify SWELL1 as a cell-autonomous sensor of adipocyte size that regulates adipocyte growth, insulin sensitivity and glucose tolerance.

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

Figure 1: Adipocyte patch clamp reveals a prominent swell-activated current in murine and human adipocytes.
Figure 2: SWELL1 mediates a swell-activated current in adipocytes.
Figure 3: SWELL1-mediated VRAC is increased in adipocytes of obese mice and humans.
Figure 4: SWELL1 regulates lipid content and glucose metabolism via insulin-pAKT2-AS160-GLUT4 and GSK3β signalling.
Figure 5: SWELL1/LRRD-GRB2-Cav1-IRS1-IR complex regulates insulin-pAKT2 signalling.
Figure 6: SWELL1 knockdown reduces adipocyte size and adiposity in obese mice.
Figure 7: Adipocyte SWELL1 is dispensable for adipose development but required for glucose homeostasis and insulin sensitivity in vivo.
Figure 8: Adipose-restricted SWELL1 deletion limits adipocyte size in obese mice.

Similar content being viewed by others

Change history

  • 22 May 2017

    In the original version of this Article, the name of author E. Dale Abel was coded wrongly, resulting in it being incorrect when exported to citation databases. This has now been corrected, though no visible changes will be apparent.

  • 25 April 2017

    In the version of this Article originally published, the axes labels 'Current (nA)' and 'Voltage (mV)' in Fig. 1h were incorrectly coloured blue. This error has been corrected in the online version of the Article.

References

  1. Farnier, C. et al. Adipocyte functions are modulated by cell size change: potential involvement of an integrin/ERK signalling pathway. Int. J. Obes. Relat. Metab. Disord. 27, 1178–1186 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Shoham, N. et al. Adipocyte stiffness increases with accumulation of lipid droplets. Biophys. J. 106, 1421–1431 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Shoham, N. et al. Static mechanical stretching accelerates lipid production in 3T3-L1 adipocytes by activating the MEK signaling pathway. Am. J. Physiol. Cell Physiol. 302, C429–C441 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Pellegrinelli, V. et al. Human adipocyte function is impacted by mechanical cues. J. Pathol. 233, 183–195 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Salans, L. B., Knittle, J. L. & Hirsch, J. The role of adipose cell size and adipose tissue insulin sensitivity in the carbohydrate intolerance of human obesity. J. Clin. Invest. 47, 153–165 (1968).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Weyer, C., Foley, J. E., Bogardus, C., Tataranni, P. A. & Pratley, R. E. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia 43, 1498–1506 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Khan, T. et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol. Cell. Biol. 29, 1575–1591 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Briand, N. et al. Caveolin-1 expression and cavin stability regulate caveolae dynamics in adipocyte lipid store fluctuation. Diabetes 63, 4032–4044 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Eduardsen, K. et al. Cell volume regulation and signaling in 3T3-L1 pre-adipocytes and adipocytes: on the possible roles of caveolae, insulin receptors, FAK and ERK1/2. Cell Physiol. Biochem. 28, 1231–1246 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Oancea, E., Wolfe, J. T. & Clapham, D. E. Functional TRPM7 channels accumulate at the plasma membrane in response to fluid flow. Circ. Res. 98, 245–253 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Numata, T., Shimizu, T. & Okada, Y. TRPM7 is a stretch- and swelling-activated cation channel involved in volume regulation in human epithelial cells. Am. J. Physiol. Cell Physiol. 292, C460–C467 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Muraki, K. et al. TRPV2 is a component of osmotically sensitive cation channels in murine aortic myocytes. Circ. Res. 93, 829–838 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Liedtke, W., Tobin, D. M., Bargmann, C. I. & Friedman, J. M. Mammalian TRPV4 (VR-OAC) directs behavioral responses to osmotic and mechanical stimuli in Caenorhabditis elegans. Proc. Natl Acad. Sci. USA 100, 14531–14536 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Spassova, M. A., Hewavitharana, T., Xu, W., Soboloff, J. & Gill, D. L. A common mechanism underlies stretch activation and receptor activation of TRPC6 channels. Proc. Natl Acad. Sci. USA 103, 16586–16591 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Coste, B. et al. Piezo proteins are pore-forming subunits of mechanically activated channels. Nature 483, 176–181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Che, H., Yue, J., Tse, H. F. & Li, G. R. Functional TRPV and TRPM channels in human preadipocytes. Pflugers Arch. 466, 947–959 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Sukumar, P. et al. Constitutively active TRPC channels of adipocytes confer a mechanism for sensing dietary fatty acids and regulating adiponectin. Circ. Res. 111, 191–200 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ye, L. et al. TRPV4 is a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis. Cell 151, 96–110 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Voss, F. K. et al. Identification of LRRC8 heteromers as an essential component of the volume-regulated anion channel VRAC. Science 344, 634–638 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Qiu, Z. et al. SWELL1, a plasma membrane protein, is an essential component of volume-regulated anion channel. Cell 157, 447–458 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kumar, L. et al. Leucine-rich repeat containing 8A (LRRC8A) is essential for T lymphocyte development and function. J. Exp. Med. 211, 929–942 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Garofalo, R. S. et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKBβ. J. Clin. Invest. 112, 197–208 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cho, H., Thorvaldsen, J. L., Chu, Q., Feng, F. & Birnbaum, M. J. Akt1/PKBα is required for normal growth but dispensable for maintenance of glucose homeostasis in mice. J. Biol. Chem. 276, 38349–38352 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Vu, B. G., Gourronc, F. A., Bernlohr, D. A., Schlievert, P. M. & Klingelhutz, A. J. Staphylococcal superantigens stimulate immortalized human adipocytes to produce chemokines. PLoS ONE 8, e77988 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Strange, K., Emma, F. & Jackson, P. S. Cellular and molecular physiology of volume-sensitive anion channels. Am. J. Physiol. 270, C711–C730 (1996).

    Article  CAS  PubMed  Google Scholar 

  26. Okada, Y., Sato, K. & Numata, T. Pathophysiology and puzzles of the volume-sensitive outwardly rectifying anion channel. J. Physiol. 587, 2141–2149 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Inoue, H., Takahashi, N., Okada, Y. & Konishi, M. Volume-sensitive outwardly rectifying chloride channel in white adipocytes from normal and diabetic mice. Am. J. Physiol. Cell Physiol. 298, C900–C909 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Syeda, R. et al. LRRC8 proteins form volume-regulated anion channels that sense ionic strength. Cell 164, 499–511 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yang, H. et al. One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell 154, 1370–1379 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fasshauer, M. et al. Essential role of insulin receptor substrate-2 in insulin stimulation of Glut4 translocation and glucose uptake in brown adipocytes. J. Biol. Chem. 275, 25494–25501 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Gonzalez, E. & McGraw, T. E. Insulin-modulated Akt subcellular localization determines Akt isoform-specific signaling. Proc. Natl Acad. Sci. USA 106, 7004–7009 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sano, H. et al. Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J. Biol. Chem. 278, 14599–14602 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Eguez, L. et al. Full intracellular retention of GLUT4 requires AS160 Rab GTPase activating protein. Cell Metab. 2, 263–272 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Medina, M. & Wandosell, F. Deconstructing GSK-3: the fine regulation of its activity. Int. J. Alzheimers Dis. 2011, 479249 (2011).

    PubMed  PubMed Central  Google Scholar 

  36. Nakae, J. et al. The forkhead transcription factor Foxo1 regulates adipocyte differentiation. Dev. Cell 4, 119–129 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Siddle, K. Molecular basis of signaling specificity of insulin and IGF receptors: neglected corners and recent advances. Front. Endocrinol. (Lausanne) 3, 34 (2012).

    Article  Google Scholar 

  38. Wary, K. K., Mariotti, A., Zurzolo, C. & Giancotti, F. G. A requirement for caveolin-1 and associated kinase Fyn in integrin signaling and anchorage-dependent cell growth. Cell 94, 625–634 (1998).

    Article  CAS  PubMed  Google Scholar 

  39. Pilch, P. F. & Liu, L. Fat caves: caveolae, lipid trafficking and lipid metabolism in adipocytes. Trends Endocrinol. Metab. 22, 318–324 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gustavsson, J. et al. Localization of the insulin receptor in caveolae of adipocyte plasma membrane. FASEB J. 13, 1961–1971 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Cohen, A. W. Caveolin-1-deficient mice show insulin resistance and defective insulin receptor protein expression in adipose tissue. Am. J. Physiol. Cell Physiol. 285, C222–C235 (2003).

    Article  CAS  PubMed  Google Scholar 

  42. Sinha, B. et al. Cells respond to mechanical stress by rapid disassembly of caveolae. Cell 144, 402–413 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sedding, D. G. et al. Caveolin-1 facilitates mechanosensitive protein kinase B (Akt) signaling in vitro and in vivo. Circ. Res. 96, 635–642 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Trouet, D. et al. Caveolin-1 modulates the activity of the volume-regulated chloride channel. J. Physiol. 520, 113–119 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Abascal, F. & Zardoya, R. LRRC8 proteins share a common ancestor with pannexins, and may form hexameric channels involved in cell–cell communication. Bioessays 34, 551–560 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Sawada, A. et al. A congenital mutation of the novel gene LRRC8 causes agammaglobulinemia in humans. J. Clin. Invest. 112, 1707–1713 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shan, X. et al. Suppression of Grb2 expression improved hepatic steatosis, oxidative stress, and apoptosis induced by palmitic acid in vitro partly through insulin signaling alteration. In Vitro Cell Dev. Biol. Anim. 49, 576–582 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Liu, X. et al. Downregulation of Grb2 contributes to the insulin-sensitizing effect of calorie restriction. Am. J. Physiol. Endocrinol. Metab. 296, E1067–E1075 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Liu, X. et al. Adipose tissue insulin receptor knockdown via a new primate-derived hybrid recombinant AAV serotype. Mol. Ther. Methods Clin. Dev. 1, 8 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  50. Metzinger, M. N. et al. Correlation of X-ray computed tomography with quantitative nuclear magnetic resonance methods for pre-clinical measurement of adipose and lean tissues in living mice. Sensors (Basel) 14, 18526–18542 (2014).

    Article  PubMed Central  Google Scholar 

  51. Lee, K. Y. et al. Lessons on conditional gene targeting in mouse adipose tissue. Diabetes 62, 864–874 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen, X. & Chalfie, M. Modulation of C. elegans touch sensitivity is integrated at multiple levels. J. Neurosci. 34, 6522–6536 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Tominaga, K. et al. The novel gene fad158, having a transmembrane domain and leucine-rich repeat, stimulates adipocyte differentiation. J. Biol. Chem. 279, 34840–34848 (2004).

    Article  CAS  PubMed  Google Scholar 

  54. Hayashi, T. et al. Factor for adipocyte differentiation 158 gene disruption prevents the body weight gain and insulin resistance induced by a high-fat diet. Biol. Pharm. Bull. 34, 1257–1263 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Cho, H. et al. Insulin resistance and a diabetes mellitus-like syndrome in mice lacking the protein kinase Akt2 (PKBβ). Science 292, 1728–1731 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Jeffery, E., Church, C. D., Holtrup, B., Colman, L. & Rodeheffer, M. S. Rapid depot-specific activation of adipocyte precursor cells at the onset of obesity. Nat. Cell Biol. 17, 376–385 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen, W. S. et al. Growth retardation and increased apoptosis in mice with homozygous disruption of the Akt1 gene. Genes. Dev. 15, 2203–2208 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Abel, E. D. Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409, 729–733 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bilal, M. Y. & Houtman, J. C. GRB2 nucleates T cell receptor-mediated LAT clusters that control PLC-gamma1 activation and cytokine production. Front. Immunol. 6, 141 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lublinsky, S., Luu, Y. K., Rubin, C. T. & Judex, S. Automated separation of visceral and subcutaneous adiposity in vivo microcomputed tomographies of mice. J. Digit. Imaging 22, 222–231 (2009).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank T. J. Jentsch (FM/MDC, Berlin) for kindly sharing SWELL1/LRRC8a antibody and A. Patapoutian for sharing Flag-SWELL1 and SWELL1Δ91/+35. RNA-Seq data presented herein were obtained at the Genomics Division of the Iowa Institute of Human Genetics. We thank R. Sigmund, J. Galbraith and M. Knudson of the University of Iowa Tissue Procurement Core facility (TPC) for services provided related to acquisition of study specimens (NCI award number P30CA086862) and Susan Walsh of the Small Animal Imaging Core, University of Iowa. We thank the University of Utah Mutation Generation and Detection Core, DNA Sequencing Core, DNA/Peptide Synthesis Core, and Transgenic Gene Targeting Mouse Facility for reagents and services. We thank M. Anderson, F. Abboud, P. Snyder, C. Benson and J. Robertson for their thoughtful review of the manuscript. We thank M. Elliot-Hudson and P. Lüken for assistance with data analysis. This work was supported by grants from the NIH NIDDK 1R01DK106009 (R.S.), the Roy J. Carver Trust (R.S.), the American Heart Association Fellow-to-Faculty Award (R.S.), an American Heart Association Postdoctoral Award (Y.Z.) and an American Cancer Society Pilot Grant (R.S.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R.S.; methodology, Y.Z., L.X., S.K.G., D.T., W.J.G., T.F., R.S.; formal analysis, R.S., Y.Z., L.X., S.K.G., D.T., A.M., W.J.G., T.F.; investigation, R.S., Y.Z., L.X., S.K.G., D.T., T.F., B.M., A.M.; resources, C.W., L.C., E.D.A., J.K.S., I.S.; writing (original draft), R.S., writing (review and editing), R.S., Y.Z., L.X., S.K.G., D.T., E.D.A.; visualization, R.S., Y.Z., L.X., S.K.G.; supervision, R.S.; funding acquisition, R.S.

Corresponding author

Correspondence to Rajan Sah.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Validation of SWELL1 knock-down. Related to Fig. 2.

(a) qRT-PCR assessment of SWELL1/LRRC8a, and LRRC8b-e mRNA in human adipocyte cell line transduced with Ad-shSWELL1-mCherry (red) compared to Ad-shSCR-mCherry (black; n = 3 culture dishes of adipocytes for each condition). (b) SWELL1 Western blot in human adipocyte cell line transduced with Ad-shSWELL1-mCherry (red) compared to Ad-shSCR-mCherry (black). β-actin serves as loading control. Representative blot from 4 independent experiments. (c) VRAC over time ± hypotonic swelling (210 mOsm) in 3T3-F442A adipocytes transduced with Ad-shSCR-mCherry (black) and Ad-shSWELL1-mCherry (red). (d) Mean peak outward (+100 mV) and peak inward (−100 mV) VRAC current density from (c) in Ad-shSCR (black, n = 5) and Ad-shSWELL1 (red, n = 4) transduced 3T3-F442A adipocytes. (e) VRAC current-voltage relationship after hypotonic swelling (210 mOsm) in human adipocytes transduced with Ad-shSCR-mCherry (black) and Ad-shSWELL1-mCherry (red). (f) Mean peak outward (+100 mV) and peak inward (−100 mV) VRAC current density from (e) in Ad-shSCR (black, n = 10) and Ad-shSWELL1 (red, n = 10) transduced human adipocytes. Uncropped blots for b are shown in Supplementary Fig. 9a–c. Significance between the indicated groups in a,d and f was calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.05), (P < 0.01).

Supplementary Figure 2 CRISPR–cas9 SWELL1 gene ablation. Related to Fig. 2.

(a) Sequences of mutant alleles showing deletions generated by CRISPR–cas9 single-guide RNA (gRNA) mediated approach (KO1). (b) CRISPR–cas9 double-gRNA mediated SWELL1 knockout (KO2) resulting in excision of the DNA fragment (240 bp) between the two cut sites. (c) PCR of the double-gRNA mediated knockout gene yields a 639 bp amplicon, reflecting the expected 240 bp deletion. (d) Schematic representation of CRISPR–cas9 mediated loxP knockin around Exon 3 to generate SWELL1fl allele. SWELL1null allele is generated by Cre-Lox mediated excision of Exon 3. (e) Cre-mediated Exon 3 deletion yields the expected PCR amplicons 426 and 196 bp from two individual primer pairs flanking loxP sites around Exon 3. The 5 Kb region between loxP sites in the SWELL1fl allele could not be amplified. Uncropped blots for c are shown in Supplementary Fig. 9a–d, and e in Supplementary Fig. 9a–e. Representative gel image from 2 independent experiments (c,e). (f) Experimental approach for generating WT and SWELL1 KO primary SVF used for differentiation into cultured primary adipocytes. (g) Bright-field image (left) of SWELL1fl primary cultured adipocytes from SVF and mCherry fluorescence image (right) showing nuclear localization of Cre-mCherry. Scale bar: 200 μm. (h) Relative mRNA expression assessed by qPCR (n = 3 each). (ik) Representative VRAC current over time ± hypotonic swelling (i), VRAC current-voltage plots upon swelling (j), and mean peak outward (+100 mV) and inward (−100 mV) VRAC current density (k) in WT and SWELL1 primary SVF (n = 5–6 each). Significance between the indicated groups in h and k were calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.05), (P < 0.01).

Supplementary Figure 3 Lean and obese mouse body weight and patient characteristics. Related to Fig. 3.

(a) Body weights of lean and obese mice from which mature adipocytes were isolated for patch-clamp recordings in Fig. 3c–h. (b) Relative mRNA expression of SWELL1/LRRC8A and LRRC8B through E assessed by qPCR in mice raised on normal chow (n = 8) compared to HFD (n = 7). (c) Age, Gender and BMI of patients from which visceral adipocytes were isolated for patch-clamp recordings in Fig. 3i–n. BMI: Body mass index. Significance between the indicated groups in a and b were calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.05), (P < 0.01), (P < 0.001).

Supplementary Figure 4 SWELL1 is required for lipogenesis and adipocyte glucose metabolism, Related to Fig. 4.

(a) Heat map displaying 2-way hierarchical clustering and mRNA expression levels of WT (n = 3) and CRISPR–cas9 SWELL1 KO (n = 3) 3T3-F442A adipocytes. Columns WT 1-3 and KO 1-3 represent RNA from individual experiments. (b) Gene Set Enrichment Analysis revealing pathways and processes negatively enriched in SWELL1 KO compared with WT 3T3-F442A adipocytes. Q value is the false discovery rate (FDR)-adjusted p value. Q < 0.10 is considered statistically significant. (c) Representative fluorescence images of AdipoRed stained WT and SWELL1 KO 3T3-F442A adipocytes under low glucose (5.6 mM) and high glucose (25 mM) culture conditions. Mean AdipoRed fluorescence intensity per cell under each condition (nWT,5.6 mM = 444, nWT,25 mM = 422, nKO,5.6 mM = 527, nKO,25 mM = 589). Measurements pooled from 3 separate independent experiments. Scale bar: 200 μm. (d) Transmission Electron Microscopy (TEM) images of glycogen granules in WT and SWELL1 KO adipocytes. Scale bar: 0.5 μm. Significance between the indicated groups in c were calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.001).

Supplementary Figure 5 SWELL1 interacts with IR/IRS1 via GRB2, Related to Fig. 5.

(a) GRB2 immunoprecipitation from 3T3-F442A adipocytes and immunoblot with insulin receptor (IR) and GRB2 antibodies. Representative blot of 3 independent experiments. (b) IR immunoprecipitation from HEK293 cells and immunoblot with GRB2 antibody. Representative blot of 3 independent experiments. (c) GRB2 immunoprecipitation from HEK293 cells ± transfection with Myc-HA-tagged insulin receptor substrate 1 (IRS-1-HA-Myc) and immunoblot with HA and GRB2 antibodies. Representative blot of 1 replicate. Uncropped blots for a are shown in Supplementary Fig. 9a, f; for b are shown in Supplementary Fig. 9a, g; for c are shown in Supplementary Fig. 9a, h.

Supplementary Figure 6 Long-term SWELL1 knock-down using AAV/Rec2, Related to Fig. 6.

(a) mCherry fluorescence indicative of AAV/Rec2 transduction at different regions of iWAT and eWAT in AAV-shRNA-mCherry injected mouse. (i) indicates the injection site. Scale bar: 200 μm. (b) mCherry fluorescence indicating AAV/Rec2 transduction in skeletal muscle (tibalis anterior) and liver in AAV-shRNA-mCherry injected mouse. Scale bar: 200 μm. mCherry fluorescence indicating AAV/Rec2 transduction at regions in a,b were visualized from 6 independent experiments. (c) Nuclear Magnetic Resonance (NMR) measurements of percent fat in AAV-shSCR-mCherry (n = 6) and AAV-shSWELL1-mCherry (n = 6) transduced mice. (d) NMR measurements of percent lean mass in AAV-shSCR-mCherry (n = 6) and AAV-shSWELL1-mCherry (n = 6) transduced mice. Significance between the indicated groups in c and d were calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.05).

Supplementary Figure 7 WT and Adipo KO adipocyte size, expression of adipocyte differentiation genes and indirect calorimetry data in lean, regular chow fed mice, Related to Fig. 7.

(a) Bright field images of mature adipocytes isolated from iWAT of lean littermate WT and Adipo KO mice. Scale bar: 50 μm. (bc) Mean adipocyte area (b) and size distribution (c) of WT (n = 1214) and Adipo KO (n = 982) adipocytes. Measurements pooled from 3 pairs of mice. (d) H&E staining of iWAT adipose tissues in lean littermate WT and Adipo KO mice. Scale bar: 50 μm. (e,f) Mean adipocyte cross-sectional area (e) and size distribution (f) of WT (n = 979) and Adipo KO (n = 1118) adipocytes. Measurements pooled from 3 pairs of mice. (g) Relative mRNA expression of adipocyte differentiation markers assessed by qPCR (nWT = 8, nAdipoKO = 5). (h-n) Metabolic cage studies were performed on WT (n = 9) and Adipo KO (n = 7) mice fed a regular chow diet (13–19 weeks of age). The mice were housed individually in the chambers for 5 days, and data was analyzed from the last 3 days. (h) Heat production; (i) Respiratory exchange ratio (RER); (j) CO2 production; (k) O2 consumption; (l) Physical activity; (m) Food consumption; and (n) Sleep. Significance between the indicated groups were calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.05).

Supplementary Figure 8 WT and Adipo KO adipocyte size and indirect calorimetry data in obese, HFD fed mice, Related to Fig. 7.

(a) Bright field images of mature primary adipocytes freshly isolated from iWAT of WT and Adipo KO mice (17 weeks HFD). (b,c) Mean adipocyte area (b) and size distribution (c) of WT (n = 284) and Adipo KO adipocytes (n = 470) in a. Measurements pooled from 3 pairs of mice. (dj) Metabolic cage studies were performed in WT (n = 7) and Adipo KO (n = 9) mice (HFD for 12–16 weeks). The mice were housed individually in the chambers for 5 days, and data was analyzed from the last 3 days. (d) Heat production; (e) Respiratory exchange ratio (RER); (f) CO2 production; (g) O2 consumption; (h) Physical activity; (i) Food consumption; and (j) Sleep. Significance between the indicated groups in a and b were calculated using a two-tailed Student’s t-test. Exact P-values are listed in Supplementary Table 6. Error bars represent mean ± s.e.m.(P < 0.05), (P < 0.001). Scale bar in a: 50 μm.

Supplementary information

Supplementary Information

Supplementary Information (PDF 51013 kb)

Supplementary Table 1

Supplementary Information (XLSX 1546 kb)

Supplementary Table 2

Supplementary Information (XLSX 21 kb)

Supplementary Table 3

Supplementary Information (XLSX 9 kb)

Supplementary Table 4

Supplementary Information (XLSX 8 kb)

Supplementary Table 5

Supplementary Information (XLSX 9 kb)

Supplementary Table 6

Supplementary Information (XLSX 13 kb)

Supplementary Table 7

Supplementary Information (XLSX 13 kb)

41556_2017_BFncb3514_MOESM102_ESM.avi

MicroCT derived 3D reconstruction of visceral adipose of representative AAV/Rec2-shSCR-mCherry transduced mouse. (AVI 3788 kb)

41556_2017_BFncb3514_MOESM103_ESM.avi

MicroCT derived 3D reconstruction of visceral adipose of representative AAV/Rec2-shSWELL1-mCherry transduced mouse. (AVI 3685 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Y., Xie, L., Gunasekar, S. et al. SWELL1 is a regulator of adipocyte size, insulin signalling and glucose homeostasis. Nat Cell Biol 19, 504–517 (2017). https://doi.org/10.1038/ncb3514

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3514

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