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:

TP53INP2 regulates adiposity by activating β-catenin through autophagy-dependent sequestration of GSK3β

Abstract

Excessive fat accumulation is a major risk factor for the development of type 2 diabetes mellitus and other common conditions, including cardiovascular disease and certain types of cancer. Here, we identify a mechanism that regulates adiposity based on the activator of autophagy TP53INP2. We report that TP53INP2 is a negative regulator of adipogenesis in human and mouse preadipocytes. In keeping with this, TP53INP2 ablation in mice caused enhanced adiposity, which was characterized by greater cellularity of subcutaneous adipose tissue and increased expression of master adipogenic genes. TP53INP2 modulates adipogenesis through autophagy-dependent sequestration of GSK3β into late endosomes. GSK3β sequestration was also dependent on ESCRT activity. As a result, TP53INP2 promotes greater β-catenin levels and induces the transcriptional activity of TCF/LEF transcription factors. These results demonstrate a link between autophagy, sequestration of GSK3β into late endosomes and inhibition of adipogenesis in vivo.

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: TP53INP2 regulates adipogenesis.
Fig. 2: TP53INP2 loss of function promotes adipose hyperplasia.
Fig. 3: TP53INP2 loss of function promotes adiposity.
Fig. 4: TP53INP2 increases WNT signalling.
Fig. 5: TP53INP2 enhances β-catenin levels.
Fig. 6: TP53INP2 facilitates the sequestration of GSK3β into a late-endosomal compartment in an ESCRT-dependent manner.
Fig. 7: Autophagy activity is necessary for the TP53INP2-dependent sequestration of GSK3β.

Similar content being viewed by others

References

  1. Wu, H. & Ballantyne, C. M. Inflammation versus host defense in obesity. Cell Metab. 20, 708–709 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Apovian, C. M. The clinical and economic consequences of obesity. Am. J. Manag. Care 19, s219–s228 (2013).

    Google Scholar 

  3. Sun, K., Kusminski, C. M. & Scherer, P. E. Adipose tissue remodeling and obesity. J. Clin. Invest. 121, 2094–2101 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gustafson, B., Hedjazifar, S., Gogg, S., Hammarstedt, A. & Smith, U. Insulin resistance and impaired adipogenesis. Trends Endocrinol. Metab. 26, 193–200 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Lundgren, M. et al. Fat cell enlargement is an independent marker of insulin resistance and ‘hyperleptinaemia’. Diabetologia 50, 625–633 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. van Tienen, F. H. et al. Preadipocytes of type 2 diabetes subjects display an intrinsic gene expression profile of decreased differentiation capacity. Int. J. Obes. 35, 1154–1164 (2011).

    Article  CAS  Google Scholar 

  7. Gustafson, B., Hammarstedt, A., Hedjazifar, S. & Smith, U. Restricted adipogenesis in hypertrophic obesity: the role of WISP2, WNT, and BMP4. Diabetes 62, 2997–3004 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hammarstedt, A. et al. WISP2 regulates preadipocyte commitment and PPARγ activation by BMP4. Proc. Natl Acad. Sci. USA 110, 2563–2568 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Patel, P. & Abate, N. Body fat distribution and insulin resistance. Nutrients 5, 2019–2027 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Spalding, K. L. et al. Dynamics of fat cell turnover in humans. Nature 453, 783–787 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Cawthorn, W. P., Scheller, E. L. & MacDougald, O. A. Adipose tissue stem cells: the great WAT hope. Trends Endocrinol. Metab. 23, 270–277 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rosen, E. D. & Spiegelman, B. M. What we talk about when we talk about fat. Cell 156, 20–44 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Rosen, E. D. & MacDougald, O. A. Adipocyte differentiation from the inside out. Nat. Rev. Mol. Cell Biol. 7, 885–896 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Christodoulides, C., Lagathu, C., Sethi, J. K. & Vidal-Puig, A. Adipogenesis and WNT signalling. Trends Endocrinol. Metab. 20, 16–24 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Jin, T. Current understanding on role of the Wnt signaling pathway effector TCF7L2 in glucose homeostasis. Endocr. Rev. 37, 254–277 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Ross, S. E. et al. Inhibition of adipogenesis by Wnt signaling. Science 289, 950–953 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Bennett, C. N. et al. Regulation of Wnt signaling during adipogenesis. J. Biol. Chem. 277, 30998–31004 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Bennett, C. N., Hodge, C. L., MacDougald, O. A. & Schwartz, J. Role of Wnt10b and C/EBPα in spontaneous adipogenesis of 243 cells. Biochem. Biophys. Res. Commun. 302, 12–16 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Cawthorn, W. P. et al. Wnt6, Wnt10a and Wnt10b inhibit adipogenesis and stimulate osteoblastogenesis through a β-catenin-dependent mechanism. Bone 50, 477–489 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Longo, K. A. et al. Wnt10b inhibits development of white and brown adipose tissues. J. Biol. Chem. 279, 35503–35509 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Wright, W. S. et al. Wnt10b inhibits obesity in ob/ob and agouti mice. Diabetes 56, 295–303 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Aslanidi, G. et al. Ectopic expression of Wnt10b decreases adiposity and improves glucose homeostasis in obese rats. Am. J. Physiol. Endocrinol. Metab. 293, E726–E736 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Singh, R. et al. Autophagy regulates adipose mass and differentiation in mice. J. Clin. Invest. 119, 3329–3339 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Baerga, R., Zhang, Y., Chen, P. H., Goldman, S. & Jin, S. Targeted deletion of autophagy-related 5 (atg5) impairs adipogenesis in a cellular model and in mice. Autophagy 5, 1118–1130 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ro, S. H. et al. Distinct functions of Ulk1 and Ulk2 in the regulation of lipid metabolism in adipocytes. Autophagy 9, 2103–2114 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang, Y. et al. Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis. Proc. Natl Acad. Sci. USA 106, 19860–19865 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kaur, J. & Debnath, J. Autophagy at the crossroads of catabolism and anabolism. Nat. Rev. Mol. Cell Biol. 16, 461–472 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Mauvezin, C. et al. The nuclear cofactor DOR regulates autophagy in mammalian and Drosophila cells. EMBO Rep. 11, 37–44 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Sancho, A. et al. DOR/Tp53inp2 and Tp53inp1 constitute a metazoan gene family encoding dual regulators of autophagy and transcription. PLoS ONE 7, e34034 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sala, D. et al. Autophagy-regulating TP53INP2 mediates muscle wasting and is repressed in diabetes. J. Clin. Invest. 124, 1914–1927 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Nowak, J. et al. The TP53INP2 protein is required for autophagy in mammalian cells. Mol. Biol. Cell 20, 870–881 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ruzankina, Y. et al. Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem Cell 1, 113–126 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ye, R. et al. Impact of tamoxifen on adipocyte lineage tracing: inducer of adipogenesis and prolonged nuclear translocation of Cre recombinase. Mol. Metab. 4, 771–778 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sanchez-Gurmaches, J. et al. PTEN loss in the Myf5 lineage redistributes body fat and reveals subsets of white adipocytes that arise from Myf5 precursors. Cell Metab. 16, 348–362 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gustafson, B. et al. Inflammation and impaired adipogenesis in hypertrophic obesity in man. Am. J. Physiol. Endocrinol. Metab. 297, E999–E1003 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Liu, C. et al. Control of β-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 108, 837–847 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Okamura, M. et al. COUP-TFII acts downstream of Wnt/β-catenin signal to silence PPARγ gene expression and repress adipogenesis. Proc. Natl Acad. Sci. USA 106, 5819–5824 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Taelman, V. F. et al. Wnt signaling requires sequestration of glycogen synthase kinase 3 inside multivesicular endosomes. Cell 143, 1136–1148 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Vinyoles, M. et al. Multivesicular GSK3 sequestration upon Wnt signaling is controlled by p120-catenin/cadherin interaction with LRP5/6. Mol. Cell 53, 444–457 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Stenmark, H. et al. Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis. EMBO J. 13, 1287–1296 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Christ, L., Raiborg, C., Wenzel, E. M., Campsteijn, C. & Stenmark, H. Cellular functions and molecular mechanisms of the ESCRT membrane-scission machinery. Trends Biochem. Sci. 42, 42–56 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Maris, M. et al. Deletion of C/EBP homologous protein (Chop) in C57Bl/6 mice dissociates obesity from insulin resistance. Diabetologia 55, 1167–1178 (2012).

    Article  CAS  PubMed  Google Scholar 

  43. Perreault, M. & Marette, A. Targeted disruption of inducible nitric oxide synthase protects against obesity-linked insulin resistance in muscle. Nat. Med. 7, 1138–1143 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Fan, W. et al. Androgen receptor null male mice develop late-onset obesity caused by decreased energy expenditure and lipolytic activity but show normal insulin sensitivity with high adiponectin secretion. Diabetes 54, 1000–1008 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Kalupahana, N. S. et al. Eicosapentaenoic acid prevents and reverses insulin resistance in high-fat diet-induced obese mice via modulation of adipose tissue inflammation. J. Nutr. 140, 1915–1922 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Cariou, B., Charbonnel, B. & Staels, B. Thiazolidinediones and PPARγ agonists: time for a reassessment. Trends Endocrinol. Metab. 23, 205–215 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Odegaard, J. I. et al. Macrophage-specific PPARγ controls alternative activation and improves insulin resistance. Nature 447, 1116–1120 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bissig, C. & Gruenberg, J. ALIX and the multivesicular endosome: ALIX in Wonderland. Trends Cell Biol. 24, 19–25 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Scourfield, E. J. & Martin-Serrano, J. Growing functions of the ESCRT machinery in cell biology and viral replication. Biochem. Soc. Trans. 45, 613–634 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Smith, U. et al. Thiazolidinediones (PPARγ agonists) but not PPARα agonists increase IRS-2 gene expression in 3T3-L1 and human adipocytes. FASEB J. 15, 215–220 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Chacon, M. R. et al. Expression of TWEAK and its receptor Fn14 in human subcutaneous adipose tissue. Relationship with other inflammatory cytokines in obesity. Cytokine 33, 129–137 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Baumgartner, B. G. et al. Identification of a novel modulator of thyroid hormone receptor-mediated action. PLoS ONE 2, e1183 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Gentleman, R. C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Irizarry, R. A. et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003).

    Article  PubMed  Google Scholar 

  55. Rossell, D., Guerra, R. & Scott, C. Semi-parametric differential expression analysis via partial mixture estimation. Stat. Appl. Genet. Mol. Biol. 7, 1544–6115 (2008).

    Article  Google Scholar 

  56. Munoz, P. et al. Expression and insulin-regulated distribution of caveolin in skeletal muscle. Caveolin does not colocalize with GLUT4 in intracellular membranes. J. Biol. Chem. 271, 8133–8139 (1996).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Rossell (Department of Statistics, University of Warwick, Coventry, UK) and C. Stephan-Otto Attolini (Biostatistics and Bioinformatics Unit, Institute for Research in Biomedicine, Barcelona, Spain) for their support with the transcriptomic studies. We also thank L. Bardia (Advanced Digital Microscopy Facility, Institute for Research in Biomedicine, Barcelona, Spain), J. Comas (Cytometry and Genomics Facility, University of Barcelona, Spain), N. Plana, V. Hernández and J. Manuel Seco for their technological assistance. A.Z. is a recipient of an ICREA 'Academia' (Generalitat de Catalunya), M.R. is a researcher hired by CIBERDEM (Instituto de Salud Carlos III) and A.S.-P. is a recipient of a predoctoral fellowship from the Universitat de Barcelona. This study was supported by research grants from the MINECO (SAF2013-40987R), grant 2014SGR48 from the Generalitat de Catalunya, CIBERDEM ('Instituto de Salud Carlos III') and INTERREG IV-B-SUDOE-FEDER (DIOMED, SOE1/P1/E178). IRB Barcelona is the recipient of a Severo Ochoa Award of Excellence from MINECO (Government of Spain). IRB Barcelona is a member of CERCA.

Author information

Authors and Affiliations

Authors

Contributions

M.R. conceived and performed the experiments and wrote the manuscript. V.A.F. conceived and performed the experiments. A.S.-P., I.C.-R. A.D.-R., X.D., B.G., A.H., M.S.-F. and J.M.M.-N. performed the experiments. J.V., J.M.F.-R. and U.S. revised the experimental data and contributed to the discussion. M.P. contributed to the discussion. A.Z. directed the research, revised the experimental data and wrote the manuscript.

Corresponding author

Correspondence to Antonio Zorzano.

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.

Integrated supplementary information

Supplementary Figure 1 TP53INP2 regulates adipogenesis.

Panels a to h: Control (C) and TP53INP2 knockdown (KD) 3T3-L1 cells. (a) TP53INP2 mRNA (n = 4 independent experiments) and protein (n = 3 independent experiments) expression. Data are mean ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test (*P < 0.0001); (b) Distribution of adipocyte cell area at day 8 of differentiation. Data are mean ± SEM. Statistical significance was calculated using two-way ANOVA (*P = 0.0005); (c) Triglycerides content. Data are mean ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test (*P < 0.0001); Panels d and e: GLUT4 (d) and perilipin (e) protein quantification in adipocytes. Data are mean ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test (GLUT4: *P = 0.0004 and Perilipin: *P < 0.0001); (f) Insulin-stimulated 2-deoxy-glucose uptake. Data are mean ± SEM (n = 6 independent experiments). Statistical significance was calculated using two-way ANOVA followed by Sidak’s test (*P = 0.0080); Panels g and h: PPARγ (g) and C/EBPα (h) protein quantification during differentiation. Data are mean ± SEM. Statistical significance was calculated using two-way ANOVA followed by Sidak’s test (PPARγ: *P < 0.0001; C/EBPα: *P = 0.0001 and #P < 0.0001); Panels i to n: Control (-) and TP53INP2- (+) overexpressing 3T3-L1CAR∆1 cells. (i) TP53INP2 mRNA and protein expression. Data are mean ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test (*P < 0.0001); (j) Triglycerides content. Data are mean ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0034); Panels k and l: GLUT-4 (k) and perilipin (l) protein quantification in adipocytes. Data are mean ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test (GLUT4: *P = 0.0125 and Perilipin: *P < 0.0496); Panels m and n: PPARγ (m) and C/EBPα (n) protein quantification during differentiation. Data are mean ± SEM. Statistical significance was calculated using two-way ANOVA followed by Sidak’s test (PPARγ: *P = 0.0049, #P = 0.0004 and §P < 0.0001; C/EBPα: *P = 0.0029 and #P = 0.0060); o) Control and TP53INP2-overexpressing human preadipocytes. PPARγ protein expression during human adipogenesis. Data are mean ± SEM. Statistical significance was calculated using two-way ANOVA followed by Sidak’s test (*P = 0.0269, #P = 0.0207 and §P < 0.0473). All unprocessed blots are provided in Supplementary Figure 8.

Supplementary Figure 2 TP53INP2 loss-of-function promotes adipose hyperplasia.

Panels a, b and c: TP53INP2 mRNA levels in human subcutaneous and visceral adipose tissue are inversely proportional to BMI. The sample size of the study 1 is n = 24 samples of scWAT (a) and n = 38 samples of vWAT (b). In the study 2, the sample size is n = 35 samples of scWAT. Statistical significance was calculated using a Spearman correlation analysis (r = -0.439 and P = 0.0051 (a); r = -0.567 and P = 0.0002 (b); r = -0.348 and P = 0.04 (c); d) Experimental design for the generation of TP53INP2 KOUbc mice: two-month-old mice expressing CreTP53INP2loxP/loxP and their non-expressing Cre-ERT2TP53INP2loxP/loxP control littermates were fed a tamoxifen diet. Experimental data were collected 2 months after starting the tamoxifen diet; Panels e to j: Control and TP53INP2 KOUbc four-month-old mice. Data represent means ± SEM. (e) TP53INP2 mRNA levels in ingWAT (n = 4 control and n = 5 KOUbc mice) and vWAT (n = 5 control and n = 7 KOUbc mice). Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0002 and #P < 0.0001); (f) TP53INP2 protein expression in ingWAT (n = 3) and pgWAT (n = 3) depots; (g) TP53INP2 mRNA levels in other tissues (n = 3 samples for each tissue). Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0139; #P = 0.0031; §P ≤ 0.0001 and &P = 0.0004); Panels h and i: Tissues weigh of females (h) (Brain, kidney and heart: n = 10 control and n = 15 KOUbc mice, and Gastroc, Quads and Tib: n = 5 control and n = 9 KOUbc mice) and males (i) (n = 4 control and n = 7 KOUbc mice). Data represent means ± SEM. Statistical significance was calculated using an unpaired two-tailed t-test; j) Fsp27, Perilipin, Leptin, Resistin, LPL and VEGF mRNA levels in adipose tissue. Data represent means ± SEM (n = 4 control and n = 6 KOUbc mice). Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0113; #P = 0.0320; §P < 0.0271; &P = 0.0452; °P = 0.0465 and +P < 0.0107). All unprocessed blots are provided in Supplementary Figure 8.

Supplementary Figure 3 TP53INP2 loss-of-function promotes adiposity.

Panels a to d: Control and TP53INP2 KOMyf5 mice. Data represent means ± SEM. (a) TP53INP2 mRNA levels in white adipose tissue depots of control (asWAT: n = 6; rWAT: n = 8; ingWAT: n = 6 and pgWAT: n = 4) and TP53INP2 KOMyf5 mice (asWAT: n = 6; rWAT: n = 8; ingWAT: n = 7 and pgWAT: n = 4). Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0020 and #P = 0.0001); (b) Body weight of control (n = 7) and TP53INP2 KOMyf5 (n = 9) mice; (c) Weight of asWAT (n = 7 control and n = 8 KOMyf5 mice), rWAT (n = 7 control and n = 8 KOMyf5 mice), ingWAT (n = 7 control and n = 10 KOMyf5 mice) and pgWAT (n = 7 control and n = 10 KOMyf5 mice); (d) Adipocyte area of ingWAT (n = 4 control and KOMyf5 mice) and pgWAT (n = 5 control and n = 6 KOMyf5 mice); e) Plasma insulin levels in control (n = 6) and TP53INP2 KOUbc (n = 6) four-month-old mice; f) Plasma insulin levels in control (n = 5) and TP53INP2 KOUbc (n = 5) eight-month-old mice; g) Plasma insulin levels in control (n = 5) and TP53INP2 KOUbc (n = 9) mice fed with high fat diet; Panels h to l: Control and TP53INP2 KOUbc four-month-old mice. (h) TNFα and IL-1β mRNA levels in ingWAT. Data are means ± SEM (TNFα: n = 5 control and KOUbc mice, respectively; IL-1β: n = 4 control and n = 8 KOUbc mice). Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0052 and #P = 0.0486); (i) Oxygen consumption of control (n = 6) and TP53INP2 KOUbc (n = 6) mice. Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0241); (j) Locomotor activity of control (n = 6) and TP53INP2 KOUbc (n = 6) mice; Panels k and l: Food (k) and water intake (l) of control (n = 6 females and n = 4 males) and TP53INP2 KOUbc (n = 6 females and n = 8 males) mice.

Supplementary Figure 4 TP53INP2 increases WNT signaling.

a) TOP-Flash and FOP-Flash reporter activity in control and TP53INP2-overexpressing HEK293T cells, and effects of LiCl. Data are means ± SEM of n = 3 independent experiments. Statistical significance was calculated using one-way ANOVA followed by Tukey’s test (*P = 0.0002 indicates effects caused by TP53INP2 overexpression and #P < 0.0001 indicates effects caused by LiCl; b) TOP-Flash and FOP-Flash reporter activity in control HEK293T cells and cells overexpressing wild-type, L36A/L40A or E97K/D98K mutant forms of TP53INP2, in the presence or absence of LiCl. Data are means ± SEM of n = 3 independent experiments. Statistical significance was calculated using one-way ANOVA followed by Tukey’s test (*P < 0.0001 indicates effects caused by TP53INP2 overexpression, and #P < 0.0001, §P = 0.0061, &P = 0.0004 indicates effects caused by LiCl).

Supplementary Figure 5 TP53INP2 enhances β-catenin levels.

a) Protein expression of total and active form of β-catenin in control and TP53INP2 KD 3T3-L1 preadipocytes. Data represent n = 3 independent experiments; b) Quantification of total and active forms of β-catenin in control and TP53INP2 KD 3T3-L1 preadipocytes. Statistical significance was calculated using an unpaired two-tailed t-test (β-catenin: *P = 0.0022 and Activeβ-catenin: *P = 0.0002); c) β-catenin mRNA levels in control and KD preadipocytes. Statistical significance was calculated using an unpaired two-tailed t-test (P = 0.6934); d) β-catenin protein quantification in ingWAT and pgWAT depots from control and TP53INP2 KOUbc mice. Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0094 and #P = 0.0073); Panels e and f: Quantification of total and active forms of β-catenin in control and TP53INP2-overexpressing 3T3-L1 CAR∆1 preadipocytes (n = 3 independent experiments) (panel e) or human preadipocytes (n = 3 independent experiments) (panel f). Statistical significance was calculated using an unpaired two-tailed t-test (3T3-L1 CAR∆1 preadipocytes: *P = 0.0016 and *P = 0.0041 for β-catenin and Activeβ-catenin, respectively; Human preadipocytes: *P = 0.0006 and *P = 0.0027 for β-catenin and Activeβ-catenin, respectively); Panels g and h: 3T3-L1 adipocytes differentiated in the presence or absence of WNT3a (n = 3 independent experiments). (g) Representative images at day 8 of differentiation; (h) Protein expression of total and active form of β-catenin, PPARγ and C/EBPα during the differentiation process. All unprocessed blots are provided in Supplementary Figure 8.

Supplementary Figure 6 TP53INP2 facilitates the sequestration of GSK3β into a late endosomal compartment in an ESCRT-dependent manner.

a) Immunofluorescence of TP53INP2 (blue) and endogenous CD63 (cyan) in the presence or absence of Rab5Q79L in Hela cells (n = 3 independent experiments). Scale bar, 8 μm; Panels b and c: β-catenin protein quantification in the presence of wild-type TP53INP2 (b) or mutant form W35/I38A (c). Data are means ± SEM (n = 3 independent experiments). Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0002 control vs TP53INP2-overexpressing cells); d) Cellular localization of GSK3β-RFP in the presence or absence of TP53INP2 in Rab5S34N-overexpressing HeLa cells (n = 3 independent experiments). Confocal images show TP53INP2 (cyan), EGFP-Rab5S34N (red), RFP-GSK3β (green) and DAPI (blue). Scale bar, 8 μm; e) Immunofluorescence staining shows endogenous CD63 (red), endogenous GSK3β (green) and DAPI (blue) in control and TP53INP2 KD 3T3-L1 cells (n = 3 independent experiments). Insets are enlargements of the outlined areas. White arrows show colocalization between endogenous CD63 and GSK3β. Scale bar, 8 μm and 1 μm (insets); f) Immunofluorescence staining shows endogenous EEA1 (green) and DAPI (blue) in control and TP53INP2 KD 3T3-L1 cells (n = 3 independent experiments). Scale bar, 10 μm; Panels g to i: HRS KD, VPS24 KD and control NIH-3T3 cells. (g) Protein expression of HRS and VPS24; (h) Protein expression of β-catenin (n = 5 independent experiments); (i) Quantification of β-catenin. Data represent the mean ± SEM of n = 5 independent experiments. Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0159 indicates effects caused by TP53INP2). All unprocessed blots are provided in Supplementary Figure 8.

Supplementary Figure 7 Autophagy activity is necessary for the TP53INP2-dependent sequestration of GSK3β.

a) LC3 protein abundance in 3T3-L1 cells upon incubation with increasing concentrations of chloroquine; b) Quantification of C/EBPα protein expression in chloroquine treated-3T3-L1 cells at 48h and 72h of adipogenic differentiation. Statistical significance was calculated using one-way ANOVA followed by Tukey’s test (*P = 0.0055 and #P = 0.0065 indicates effects caused by CQ treatment); Panels c and d: Atg5+/+ and Atg5-/- MEF cells. (c) β-catenin protein abundance in the presence or absence of TP53INP2; (d) β-catenin protein quantification. Data are means ± SEM of n = 3 independent experiments from control and TP53INP2-overexpressing cells. Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0079 control vs TP53INP2-overexpressing cells); Panels e and f: Atg3+/+ and Atg3-/- MEF cells. (e) Protein expression of β-catenin in the presence or absence of TP53INP2; (f) β-catenin protein quantification. Data are means ± SEM of n = 6 independent experiments from control and TP53INP2-overexpressing cells. Statistical significance was calculated using an unpaired two-tailed t-test (*P = 0.0001 control vs TP53INP2-overexpressing cells; g) Immunofluorescence of endogenous LC3-II in Rab5Q79L-overexpressing Atg5+/+ and Atg5-/- MEFs in the presence of TP53INP2. Scale bar, 8 μm. Data represent n = 3 independent experiments. Confocal images show TP53INP2 (cyan), DsRed-Rab5Q79L (red), GSK3β-GFP (green) and LC3II (blue). All unprocessed blots are provided in Supplementary Figure 8.

Supplementary Figure 8 Unprocessed images of blots.

a) Blots corresponding to the Fig. 1c; b) Blots corresponding to the Fig. 1f; c) Blots corresponding to the Fig. 1h; d) Blots corresponding to the Fig. 1k; e) Blots corresponding to the Fig. 1m. f) Blots corresponding to the Fig. 5a; g) Blots corresponding to the Fig. 5b; h) Blots corresponding to the Fig. 5c; i) Blots corresponding to the Fig. 5d; j) Blots corresponding to the Fig. 5f; k) Blots corresponding to the Fig. 5j; l) Blots corresponding to the Fig. 6c. m) Blots corresponding to the figure S1a; n) Blots corresponding to the figure S1i; o) Blots corresponding to the figure S2f; p) Blots corresponding to the figure S5a; q) Blots corresponding to the figure S5h. r) Blots corresponding to the figure S6g; s) Blots corresponding to the figure S6h; t) Blots corresponding to the figure S7a; v) Blots corresponding to the figure S7e; w) Blots corresponding to the figure S7c.

Supplementary information

Supplementary Information

Supplementary Figures 1–8 and Supplementary Table legends.

Life Sciences Reporting Summary

Supplementary Table 1

List of Wnt pathway-related genes regulated in TP53INP2 KD 3T3-L1 pre-adipocytes.

Supplementary Table 2

List of Wnt pathway-related genes regulated in ingWAT from TP53INP2 KOUbc mice.

Supplementary Table 3

List of Wnt pathway-related genes regulated in pgWAT from TP53INP2 KOUbc mice.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Romero, M., Sabaté-Pérez, A., Francis, V.A. et al. TP53INP2 regulates adiposity by activating β-catenin through autophagy-dependent sequestration of GSK3β. Nat Cell Biol 20, 443–454 (2018). https://doi.org/10.1038/s41556-018-0072-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0072-9

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