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:

The lipid-droplet-associated protein ABHD5 protects the heart through proteolysis of HDAC4

Abstract

Catecholamines stimulate the first step of lipolysis through protein kinase A (PKA)-dependent release of the lipid-droplet-associated protein abhydrolase domain containing 5 (ABHD5) from perilipin to coactivate the lipase adipose triglyceride lipase (ATGL). Here, we unmask a proteolytic and cardioprotective function of ABHD5. ABHD5 acts in vivo and in vitro as a serine protease that cleaves histone deacetylase 4 (HDAC4). Through the production of an amino-terminal polypeptide of HDAC4 (HDAC4-NT), ABHD5 inhibits MEF2-dependent gene expression and thereby controls glucose handling. ABHD5 deficiency leads to neutral-lipid storage disease in mice. Cardiac-specific gene therapy using the gene encoding HDAC4-NT does not protect against intracardiomyocyte lipid accumulation, but strikingly protects against heart failure, thereby challenging the concept of lipotoxicity-induced heart failure. ABHD5 levels are reduced in failing human hearts, and murine transgenic ABHD5 expression protects against pressure-overload-induced heart failure. These findings represent a conceptual advance by connecting lipid with glucose metabolism through HDAC4 proteolysis, and enable new translational approaches to treating cardiometabolic disease.

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: ABHD5 is required for HDAC4 proteolysis.
Fig. 2: ABHD5 possesses intrinsic serine protease activity.
Fig. 3: HDAC4-NT protects against heart failure in NLSD.
Fig. 4: ABHD5 protects against heart failure.
Fig. 5: Working model.

Similar content being viewed by others

Data availability

Raw data from experiments presented in Fig. 3f,g, Supplementary Table 1 and Extended Data Fig. 3d were deposited in the gene-expression omnibus (GEO), accession number GSE135662. Source Data are available for Figs. 14 and Extended Data Figs. 16, including all uncut gels of the immunoblotting figures in the manuscript, and all data supporting the findings of the manuscript can be obtained from the corresponding author upon request.

References

  1. Lohse, M. J., Engelhardt, S. & Eschenhagen, T. What is the role of beta-adrenergic signaling in heart failure? Circ. Res. 93, 896–906 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Dewenter, M., von der Lieth, A., Katus, H. A. & Backs, J. Calcium signaling and transcriptional regulation in cardiomyocytes. Circ. Res. 121, 1000–1020 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Rockman, H. A., Koch, W. J. & Lefkowitz, R. J. Cardiac function in genetically engineered mice with altered adrenergic receptor signaling. Am. J. Physiol. 272, H1553–H1559 (1997).

    CAS  PubMed  Google Scholar 

  4. Wang, H. et al. Unique regulation of adipose triglyceride lipase (ATGL) by perilipin 5, a lipid droplet-associated protein. J. Biol. Chem. 286, 15707–15715 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yamaguchi, T. et al. CGI-58 facilitates lipolysis on lipid droplets but is not involved in the vesiculation of lipid droplets caused by hormonal stimulation. J. Lipid Res. 48, 1078–1089 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Sahu-Osen, A. et al. CGI-58/ABHD5 is phosphorylated on Ser239 by protein kinase a: control of subcellular localization. J. Lipid Res. 56, 109–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Haemmerle, G. et al. ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-alpha and PGC-1. Nat. Med. 17, 1076–1085 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zierler, K. A. et al. Functional cardiac lipolysis in mice critically depends on comparative gene identification-58. J. Biol. Chem. 288, 9892–9904 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pollak, N. M. et al. The interplay of protein kinase A and perilipin 5 regulates cardiac lipolysis. J. Biol. Chem. 290, 1295–1306 (2015).

    Article  PubMed  CAS  Google Scholar 

  10. Lass, A. et al. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI-58 and defective in chanarin-dorfman syndrome. Cell Metab. 3, 309–319 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Cerk, I. K., Wechselberger, L. & Oberer, M. Adipose triglyceride lipase regulation: an overview. Curr. Protein Pept. Sci. 19, 221–233 (2018).

    CAS  PubMed  Google Scholar 

  12. Yamaguchi, T. Crucial role of CGI-58/alpha/beta hydrolase domain-containing protein 5 in lipid metabolism. Biol. Pharm. Bull. 33, 342–345 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Chanarin, I. et al. Neutral-lipid storage disease: a new disorder of lipid metabolism. BMJ 1, 553–555 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lefevre, C. et al. Mutations in CGI-58, the gene encoding a new protein of the esterase/lipase/thioesterase subfamily, in chanarin-dorfman syndrome. Am. J. Hum. Genet. 69, 1002–1012 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Radner, F. P. et al. Growth retardation, impaired triacylglycerol catabolism, hepatic steatosis, and lethal skin barrier defect in mice lacking comparative gene identification-58 (CGI-58). J. Biol. Chem. 285, 7300–7311 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Wang, W. et al. Sustained β1-adrenergic stimulation modulates cardiac contractility by Ca2+/calmodulin kinase signaling pathway. Circ. Res. 95, 798–806 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Fischer, T. H. et al. Ca2+/calmodulin-dependent protein kinase II and protein kinase A differentially regulate sarcoplasmic reticulum Ca2+ leak in human cardiac pathology. Circulation 128, 970–981 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Backs, J., Song, K., Bezprozvannaya, S., Chang, S. & Olson, E. N. CaM kinase II selectively signals to histone deacetylase 4 during cardiomyocyte hypertrophy. J. Clin. Invest. 116, 1853–1864 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Backs, J. et al. Selective repression of MEF2 activity by PKA-dependent proteolysis of HDAC4. J. Cell Biol. 195, 403–415 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lehmann, L. H. et al. A proteolytic fragment of histone deacetylase 4 protects the heart from failure by regulating the hexosamine biosynthetic pathway. Nat. Med. 24, 62–72 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Kronlage, M. et al. O-GlcNAcylation of histone deacetylase 4 protects the diabetic heart from failure. Circulation 140, 580–594 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Kim, Y. et al. The MEF2D transcription factor mediates stress-dependent cardiac remodeling in mice. J. Clin. Invest. 118, 124–132 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Yang, L. K. & Tao, Y. X. Physiology and pathophysiology of the β3-adrenergic receptor. Prog. Mol. Biol. Transl. Sci. 161, 91–112 (2019).

    Article  PubMed  CAS  Google Scholar 

  24. Wei, J. et al. Reversal of pathological cardiac hypertrophy via the MEF2-coregulator interface. JCI Insight 2, pii: 91068 (2017).

    Article  Google Scholar 

  25. Pouleur, A. C. et al. Rationale and design of a multicentre, randomized, placebo-controlled trial of mirabegron, a β3-adrenergic receptor agonist on left ventricular mass and diastolic function in patients with structural heart disease β3-left ventricular hypertrophy (β3-LVH). ESC Heart Fail. 5, 830–841 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang, Y. et al. Adipocyte liver kinase b1 suppresses beige adipocyte renaissance through class iia histone deacetylase 4. Diabetes 66, 2952–2963 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sohal, D. S. et al. Temporally regulated and tissue-specific gene manipulations in the adult and embryonic heart using a tamoxifen-inducible Cre protein. Circ. Res. 89, 20–25 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. deAlmeida, A. C., van Oort, R. J. & Wehrens, X. H. Transverse aortic constriction in mice. J. Vis. Exp. 38, e1729 (2010).

    Google Scholar 

  29. Kreusser, M. M. et al. Cardiac CaM kinase II genes delta and gamma contribute to adverse remodeling but redundantly inhibit calcineurin-induced myocardial hypertrophy. Circulation 130, 1262–1273 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Muller, O. J., Schinkel, S., Kleinschmidt, J. A., Katus, H. A. & Bekeredjian, R. Augmentation of AAV-mediated cardiac gene transfer after systemic administration in adult rats. Gene Ther. 15, 1558–1565 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Geisler, A. et al. microRNA122-regulated transgene expression increases specificity of cardiac gene transfer upon intravenous delivery of AAV9 vectors. Gene Ther. 18, 199–209 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Jungmann, A., Leuchs, B., Katus, H. A., Rommelaere, J. & Muller, O. J. Protocol for efficient generation and characterization of adeno-associated viral (AAV) vectors. Hum. Gene Ther. Methods 28, 235–246 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Lehmann, L. H. et al. Essential role of sympathetic endothelin a receptors for adverse cardiac remodeling. Proc. Natl Acad. Sci. USA 111, 13499–13504 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Adams, J. et al. 13-cis retinoic acid inhibits development and progression of chronic allograft nephropathy. Am. J. Pathol. 167, 285–298 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Benkert, P., Biasini, M. & Schwede, T. Toward the estimation of the absolute quality of individual protein structure models. Bioinformatics 27, 343–350 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Bienert, S. et al. The SWISS-MODEL repository-new features and functionality. Nucleic Acids Res. 45, D313–D319 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Waterhouse, A. et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 46, W296–W303 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Guex, N., Peitsch, M. C. & Schwede, T. Automated comparative protein structure modeling with SWISS-MODEL and Swiss-PdbViewer: a historical perspective. Electrophoresis 30 (Suppl. 1), S162–S173 (2009).

    Article  PubMed  Google Scholar 

  39. Boeszoermenyi, A. et al. Structure of a CGI-58 motif provides the molecular basis of lipid droplet anchoring. J. Biol. Chem. 290, 26361–26372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Badin, P. M. et al. Regulation of skeletal muscle lipolysis and oxidative metabolism by the co-lipase CGI-58. J. Lipid Res. 53, 839–848 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Poschl, J. M. et al. Effects of dietary supplementation of saturated fatty acids and of n-6 or n-3 polyunsaturated fatty acids on plasma and red blood cell membrane phospholipids and deformability in weanling guinea pigs. Lipids 34, 467–473 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Dodt, M., Roehr, J. T., Ahmed, R. & Dieterich, C. FLEXBAR-flexible barcode and adapter processing for next-generation sequencing platforms. Biology (Basel) 1, 895–905 (2012).

    Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Alexa, A., Rahnenfuhrer, J. & Lengauer, T. Improved scoring of functional groups from gene expression data by decorrelating GO graph structure. Bioinformatics 22, 1600–1607 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Hagenmüller for help with figures and manuscript editing; Q. Sun, M. Oestringer, J. Krebs, U. Oehl and J. Hartmann for technical help; R. Zahn, K. Perez, K. Remans and J. Baßler for help with purification of recombinant HDAC4 and ABHD5; and S. Kaden for the help with electron microscopy. L.H.L. is recipient of the Clinician-Scientist Program (CSP) of the German Cardiac Society (DGK). M.O. is supported by the Austrian Science Fund (FWF): F73 SFB Lipid Hydrolysis. J.B. was supported by grants from the Deutsche Forschungsgemeinschaft (BA 2258/2-1BA; BA 2258/9-1; SFB 1118), the European Commission (FP7-Health-2010; MEDIA-261409) and the DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung—German Centre for Cardiovascular Research) and by the BMBF (German Ministry of Education and Research).

Author information

Authors and Affiliations

Authors

Contributions

J.B. and Z.H.J. designed the study. Z.H.J., S.K.D., J.T., M.D., C.X., F.S., D.S., X.G., L.W. B.C.W., G.F., and S.W.S. carried out experiments. Z.H.J., L.H.L., S.W.S., C.D, J.-H.G. and J.B. analysed and interpreted data. T.F, O.J.M, S.S., P.M., C.D, C.M., M.O., G.H, H.A.K., J.T., and J.B. provided research support and conceptual advice. Z.H.J. J.T. and J.B. wrote the paper.

Corresponding author

Correspondence to Johannes Backs.

Ethics declarations

Competing interests

Z.H.J., L.H.L, O.J.M, H.A.K. and J.B. filed a patent on HDAC4-NT and ABHD5 gene therapy (US9914912B2). All other authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt.

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

Extended data

Extended Data Fig. 1 ABHD5 is required for HDAC4 proteolysis.

a, Immunoblotting using an antibody directed against endogenous ABHD5 in cardiac and cellular COS as well as HEK293 extracts. GAPDH was used as loading control. b, Cytosolic and nuclear fractions from adult rat ventricular myocytes were used for immunoblotting using an antibody directed against ABHD5, GAPDH and histone H3. a,b, Representative results of two independent experiments. c, Left, Immunoblotting analysis of cardiac extracts of control (Ctrl)- or isoproterenol (ISO)-treated mice using antibodies directed against HDAC4, ABHD5 and GAPDH as loading control. Middle left, Densitometric analysis of HDAC4-NT/FL ratio shown as fold change versus control. Middle right and right, Quantification of cardiac ABHD5 protein normalized to GAPDH and Abhd5 RNA levels normalized to 18S in unstressed mice and ISO-treated mice. Statistical analysis: Values are presented as mean ± s.e.m., n = 3 (n represents mice per indicated group); by unpaired two tailed t-test, P<0.05 considered as significant. d, Left, Immunoblotting analysis of cardiac extracts of control (Ctrl) or CL 316,243 (CL)-treated mice using antibodies directed against HDAC4, ABHD5 and GAPDH as loading control. Middle left, Densitometric analysis of HDAC4-NT/FL ratio shown as fold change versus control. Middle and Middle Right, Quantification of cardiac ABHD5 protein normalized to GAPDH and Abhd5 RNA levels normalized to 18S in unstressed mice and CL-treated mice. Right, Quantification of Ucp1 mRNA normalized to 18S in brown adipose tissue after treatment with CL as a marker of CL effective administration. Statistical analysis: Values are presented as mean ± s.e.m., n = 3 (n represents mice per indicated group); by unpaired two tailed t-test, P < 0.05 considered as significant.

Source data

Extended Data Fig. 2 ABHD5 possesses intrinsic serine-protease activity.

a, Prediction of 3D structure of ABHD5 using SWISS-MODEL. Highlighted are aa predicted to serve as putative catalytic triad (Asn153, Ser298 and His327) and the putative phosphorylation site Ser-237. Ser-237 and Ser-298 of ABHD5 lie in spatial proximity to each other. Lys 51 represents the N-terminal starting aa of the peptide sequence used for modelling. Pink, cap domain; green, α/β-hydrolase core. b, Multiple sequence alignment of ABHD5 protein of different species as indicated by UniProt online tool (https://uniprot.org/align/). Red boxes denote aa N153, S298 and H327 of the predicted serine-protease catalytic triad, green box denotes aa S237 as reported PKA phosphorylation site (aa numbering corresponds to human ABHD5 protein). c, Tryptophan fluorescence decay plot upon thermal denaturation of rec-ABHD5-WT, -N153D, -S237A, -S237E -S298A and -H327A (1 °C min–1). Left, The ratio of fluorescence at 350 nm/330 nm was plotted against the temperature. Right, Equivalent analysis to identify the melting temperature (Tm) of the rec-proteins. Measurements were performed once. d, Coomassie staining after protease assay using recombinant purified HDAC4 (rec-HDAC4) and increasing amounts of recombinant purified ABHD5 (rec-ABHD5) as indicated. e, Coomassie staining after protease assay using rec-HDAC4 and rec-ABHD5 in the presence or absence of serine-protease-inhibitor AEBSF as indicated. f, Coomassie staining after protease assay using rec-HDAC4 and rec-ABHD5 WT or putative catalytic-triad mutants as indicated. The data in df represent results of three independent experiments. g, Immunoblotting and Coomassie staining for HDAC4 and ABHD5 after in vitro proteolysis assays involving rec-HDAC4, rec-ABHD5 WT or Ser-237 phospho-death (S237A) and phospho-mimetic (S237E) mutants as indicated. Coomassie staining was used to quantify the proteolytic activity of ABHD5. Statistical analysis: Values are presented as mean ± s.e.m., n = 3 (n represents independent experiments); by one-way ANOVA, P < 0.05 considered as significant. h, Immunoblotting for HDAC4 and ABHD5 in yeast-cell extracts after proteolysis assay in a yeast expression system as indicated. Because HDAC4 contains the Protein-A (PRA; 15 kDa) tag, HDAC4-NT is expected to migrate at 43 kDa. The experiment was performed at least three times with similar results.

Source data

Extended Data Fig. 3 HDAC4-NT protects from ABHD5-deficiency induced cardiac dysfunction.

Cre-expressing control mice (Cre), cardiac-specific ABHD5-KO (cKO) and cKO with HDAC4-NT gene delivery (cKO+AAV9-NT) were analysed. a, Heart weight/body weight ratios (HW/BW) and fractional shortening (FS) are shown for the indicated groups. Statistical analysis: Values are presented as mean ± s.e.m.; n = 5 (n represents mice per indicated group); by one-way ANOVA, P < 0.05 considered as significant. b, Representative images of Methylene Blue staining of cardiac thin-sections to qualitatively visualize lipid droplet content in the indicated groups. Arrows point to positive lipid stain. c, Representative electron-microscopic images of the mitochondrial and sarcomeric structures of cardiomyocytes of control mice (Cre) and cKO mice; arrows point to the lipid accumulation on the mitochondrial and sarcomeric surface in cKO. b,c, Representative samples of one experiment with 5 mice included. d, RNA-seq analysis of heart tissue of the three above mentioned groups. The left panel shows a heat map displaying expression differences in cKO versus cKO+AAV9-NT. Right, Fold change in expression of selected genes in cKO versus Cre, cKO versus cKO+AAV9-NT and cKO+AAV9-NT versus Cre.

Source data

Extended Data Fig. 4 ABHD5 inhibits MEF2 and cardiomyocyte hypertrophy.

a, NRVMs were transduced with the Ad3×MEF2C-Luc reporter. Ad.FLAG–HDAC4, Ad.ABHD5 plus GFP on a bicistronic promoter and Ad.GFP (10 MOI) were then coexpressed as indicated, and NRVMs were stimulated for 24 h with ET-1 or FCS; n = 5 (n represents independent samples) and values are presented as mean ± s.e.m. b, Right panel: Fluorescence microscopy of NRVMs expressing Ad.ABHD5 plus GFP on a bicistronic promoter or Ad.GFP (10 MOI each). NRVM hypertrophy was induced with 100 nM ET-1 or 10% FCS for 24 h. Shown are representative images of α-actinin staining (red) of NRVMs and GFP fluorescence to confirm adenoviral expression. Images are representative of at least three independent experiments. Scale bar, 50 μm. Right panel, Quantification of NRVM cross-sectional areas (CSA). Statistical analysis: Values are presented as mean ± s.e.m., n = 4 (n represents independent samples); by one-way ANOVA, P < 0.05 considered as significant.

Source data

Extended Data Fig. 5 Generation and baseline characterization of cardiac-specific ABHD5 transgenic mice (TG).

a, Microinjected fragment of an expression plasmid containing hABHD5 cDNA flanked by α-MHC promoter and human growth hormone (hGH) poly-A (+) signal. b, Cardiac overexpression of ABHD5 in mice of 4 different transgenic lines as compared to wild-type littermates (WT) from the same lines was evaluated by immunoblotting analysis using an anti-ABHD5 antibody. c–e, Baseline characterization of transgenic mice. c, Representative echocardiography of a mouse from transgenic line #1 as compared to a WT littermate. The data in b are representative of 4 different mouse lines and the data in c are representative of 4 mice from line # 1 as well as 7 (WT) and 10 (TG) mice from line #9. d,e, Quantification of ejection fraction (EF), heart weight/body weight ratio (HW/BW), heart weight/tibia length ratio (HW/TL) in WT littermates (n = 4) and transgenic mice from line #1 (n = 4) (d) and in WT littermates (n = 7) and transgenic mice from line #9 (n = 10, TG) (e). d,e, Statistical analysis: Values are presented as mean ± s.e.m.; by unpaired two tailed t-test, P < 0.05 considered as significant. n represents mice per indicated group.

Source data

Extended Data Fig. 6 ABHD5 attenuates cardiac hypertrophy upon TAC.

a, Left panel: Quantification of HW/BW ratio of WT and TG mice three weeks after TAC or sham surgery. Right panel: Quantification of fractional shortening (FS) of WT and TG mice 3 weeks after TAC or sham surgery. Statistical analysis: Values are presented as mean ± s.e.m., n = 5 (n represents mice per indicated group); by one-way ANOVA, P < 0.05 considered as significant. b, mRNA expression of Abhd5 in sham and TAC-operated WT mice normalized to Gapdh. Statistical analysis: Values are presented as mean ± s.e.m., n = 4 (n represents mice per indicated group); by unpaired two tailed t-test, P < 0.05 are considered as significant. c, Corresponding band intensities of experiment shown in Fig. 4d were quantified. Left, Ratio between HDAC4-NT and HDAC4-FL. Middle, HDAC4 P-632 levels were determined upon TAC in relation to sham in WT and TG mice and normalized to GAPDH. Right, Ratio between HDAC4 P-632 and total HDAC4. Statistical analysis: Values are presented as mean ± s.e.m., n = 3 (n represents mice per indicated group); by unpaired two tailed t-test; P < 0.05 are considered as significant.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–7

Reporting Summary

Supplementary Data 1

Human serine-protease siRNA library. List of siRNAs targeting known and predicted serine proteases (library was purchased from Ambion)

Supplementary Data 2

Prediction of the 3D structure of ABHD5 extracted from the online protein-structure-homology-modelling server SWISS-MODEL (https://swissmodel.expasy.org)

Source data

Source data Fig. 1

Unprocessed Western Blots and/or gels

Source Data Fig. 1

Statistical Source Data

Source data Fig. 2

Unprocessed Western Blots and/or gels

Source data Fig. 2

Statistical Source Data

Source data Fig. 3

Unprocessed Western Blots and/or gels

Source data Fig. 3

Statistical Source Data

Source data Fig. 4

Unprocessed Western Blots and/or gels

Source data Fig. 4

Statistical Source Data

Source data Extended Data Fig. 1

Unprocessed Western Blots and/or gels

Source data Extended Data Fig. 1

Statistical Source Data

Source data Extended Data Fig. 2

Unprocessed Western Blots and/or gels

Source data Extended Data Fig. 2

Statistical Source Data

Source data Extended Data Fig. 3

Statistical Source Data

Source data Extended Data Fig. 4

Statistical Source Data

Source data Extended Data Fig. 5

Unprocessed Western Blots and/or gels

Source data Extended Data Fig. 5

Statistical Source Data

Source data Extended Data Fig. 6

Statistical Source Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jebessa, Z.H., Shanmukha, K.D., Dewenter, M. et al. The lipid-droplet-associated protein ABHD5 protects the heart through proteolysis of HDAC4. Nat Metab 1, 1157–1167 (2019). https://doi.org/10.1038/s42255-019-0138-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-019-0138-4

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