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:

A Fbxo48 inhibitor prevents pAMPKα degradation and ameliorates insulin resistance

Abstract

The adenosine monophosphate (AMP)-activated protein kinase (Ampk) is a central regulator of metabolic pathways, and increasing Ampk activity has been considered to be an attractive therapeutic target. Here, we have identified an orphan ubiquitin E3 ligase subunit protein, Fbxo48, that targets the active, phosphorylated Ampkα (pAmpkα) for polyubiquitylation and proteasomal degradation. We have generated a novel Fbxo48 inhibitory compound, BC1618, whose potency in stimulating Ampk-dependent signaling greatly exceeds 5-aminoimidazole-4-carboxamide-1-β-ribofuranoside (AICAR) or metformin. This compound increases the biological activity of Ampk not by stimulating the activation of Ampk, but rather by preventing activated pAmpkα from Fbxo48-mediated degradation. We demonstrate that, consistent with augmenting Ampk activity, BC1618 promotes mitochondrial fission, facilitates autophagy and improves hepatic insulin sensitivity in high-fat-diet-induced obese mice. Hence, we provide a unique bioactive compound that inhibits pAmpkα disposal. Together, these results define a new pathway regulating Ampk biological activity and demonstrate the potential utility of modulating this pathway for therapeutic benefit.

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: pAmpkα undergoes SCFFbxo48-mediated ubiquitin proteasomal degradation.
Fig. 2: Small-molecule Fbxo48 inhibitors increase pAmpkα.
Fig. 3: Fbxo48 inhibitor interrupts Fbxo48/pAmpkα interaction.
Fig. 4: BC1618 facilitates mitochondrial fission and autophagy.
Fig. 5: BC1618 improves hepatic insulin sensitivity in diet-induced obese mice.

Similar content being viewed by others

Data availability

All relevant data are available from the authors and/or included in the manuscript or Supplementary Information. Source data are provided with this paper. Other data that support the findings of this study are available from the corresponding author upon reasonable request. Phospho-proteomics data can be accessed at MassIVE database under ID MSV000086334.

References

  1. Hardie, D. G., Ross, F. A. & Hawley, S. A. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 13, 251–262 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mihaylova, M. M. & Shaw, R. J. The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat. Cell Biol. 13, 1016–1023 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hardie, D. G. AMPK: positive and negative regulation, and its role in whole-body energy homeostasis. Curr. Opin. Cell Biol. 33, 1–7 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Kim, J. et al. AMPK activators: mechanisms of action and physiological activities. Exp. Mol. Med. 48, e224 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sanders, M. J. et al. Investigating the mechanism for AMP activation of the AMP-activated protein kinase cascade. Biochem. J. 403, 139–148 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Herzig, S. & Shaw, R. J. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 19, 121–135 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Cool, B. et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 3, 403–416 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Myers, R. W. et al. Systemic pan-AMPK activator MK-8722 improves glucose homeostasis but induces cardiac hypertrophy. Science 357, 507–511 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Carling, D. AMPK signalling in health and disease. Curr. Opin. Cell Biol. 45, 31–37 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Steneberg, P. et al. PAN-AMPK activator O304 improves glucose homeostasis and microvascular perfusion in mice and type 2 diabetes patients.JCI Insight 3, e99114 (2018).

    Article  PubMed Central  Google Scholar 

  11. Zhou, G. et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest. 108, 1167–1174 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hawley, S. A. et al. Complexes between the LKB1 tumor suppressor, STRADα/β and MO25α/β are upstream kinases in the AMP-activated protein kinase cascade. J. Biol. 2, 28 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Shaw, R. J. et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc. Natl Acad. Sci. USA 101, 3329–3335 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Deshaies, R. J. & Joazeiro, C. A. RING domain E3 ubiquitin ligases. Annu. Rev. Biochem. 78, 399–434 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Xiu, X. et al. Genetic analysis of the FBXO48 gene in Chinese Han patients with Parkinson disease. Neurosci. Lett. 541, 224–226 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Smith, B. K. et al. Treatment of nonalcoholic fatty liver disease: role of AMPK. Am. J. Physiol. Endocrinol. Metab. 311, E730–E740 (2016).

    Article  PubMed  Google Scholar 

  17. Corton, J. M. et al. 5-Aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells? Eur. J. Biochem. 229, 558–565 (1995).

    Article  CAS  PubMed  Google Scholar 

  18. Fullerton, M. D. et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat. Med. 19, 1649–1654 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jafari, R. et al. The cellular thermal shift assay for evaluating drug target interactions in cells. Nat. Protoc. 9, 2100–2122 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Sengupta, S. et al. Discovery of NV-5138, the first selective brain mTORC1 activator. Sci. Rep. 9, 4107 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Chen, Y. et al. A small molecule NRF2 activator BC-1901S ameliorates inflammation through DCAF1/NRF2 axis. Redox Biol. 32, 101485 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Krug, K. et al. A curated resource for phosphosite-specific signature analysis. Mol. Cell Proteom. 18, 576–593 (2019).

    Article  CAS  Google Scholar 

  23. Gwinn, D. M. et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 30, 214–226 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chang, T. W. et al. Does OKT3 monoclonal antibody react with an antigen-recognition structure on human T cells? Proc. Natl Acad. Sci. USA 78, 1805–1808 (1981).

    Article  CAS  PubMed  Google Scholar 

  25. Tamas, P. et al. Regulation of the energy sensor AMP-activated protein kinase by antigen receptor and Ca2+ in T lymphocytes. J. Exp. Med. 203, 1665–1670 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zogovic, N. et al. Coordinated activation of AMP-activated protein kinase, extracellular signal-regulated kinase, and autophagy regulates phorbol myristate acetate-induced differentiation of SH-SY5Y neuroblastoma cells. J. Neurochem. 133, 223–232 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Toyama, E. Q. et al. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science 351, 275–281 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Egan, D. F. et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331, 456–461 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Kim, J. et al. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, 132–141 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mizushima, N. & Yoshimori, T. How to interpret LC3 immunoblotting. Autophagy 3, 542–545 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Hoogendijk, A. J. et al. AMP-activated protein kinase activation by 5-aminoimidazole-4-carbox-amide-1-β-d-ribofuranoside (AICAR) reduces lipoteichoic acid-induced lung inflammation. J. Biol. Chem. 288, 7047–7052 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Park, D. W. et al. Activation of AMPK enhances neutrophil chemotaxis and bacterial killing. Mol. Med. 19, 387–398 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Castanares-Zapatero, D. et al. Connection between cardiac vascular permeability, myocardial edema and inflammation during sepsis: role of the α1AMP-activated protein kinase isoform. Crit. Care Med. 41, e411–e422 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Salminen, A., Hyttinen, J. M. T. & Kaarniranta, K. AMP-activated protein kinase inhibits NF-κB signaling and inflammation: impact on healthspan and lifespan. J. Mol. Med. 89, 667–676 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ruderman, N. B. et al. AMPK, insulin resistance and the metabolic syndrome. J. Clin. Invest. 123, 2764–2772 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Viollet, B. et al. Cellular and molecular mechanisms of metformin: an overview. Clin. Sci. 122, 253–270 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  37. Grahame Hardie, D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm. Sin. B 6, 1–19 (2016).

    Article  PubMed  Google Scholar 

  38. Xiao, B. et al. Structural basis of AMPK regulation by small molecule activators. Nat. Commun. 4, 3017 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hawley, S. A. et al. The ancient drug salicylate directly activates AMP-activated protein kinase. Science 336, 918–922 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Owen, M. R., Doran, E. & Halestrap, A. P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 348, 607–614 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, C. S. et al. Metformin activates AMPK through the lysosomal pathway. Cell Metab. 24, 521–522 (2016).

    Article  PubMed  Google Scholar 

  42. Type 2 diabetes and metformin. First choice for monotherapy: weak evidence of efficacy but well-known and acceptable adverse effects. Prescrire Int 23, 269–272 (2014).

  43. Chen, W. et al. The ubiquitin E3 ligase SCF-FBXO24 recognizes deacetylated nucleoside diphosphate kinase A to enhance its degradation. Mol. Cell. Biol. 35, 1001–1013 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Zhang, C. S. et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature 548, 112–116 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pineda, C. T. et al. Degradation of AMPK by a cancer-specific ubiquitin ligase. Cell 160, 715–728 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Vlotides, G. et al. Anticancer effects of metformin on neuroendocrine tumor cells in vitro. Hormones 13, 498–508 (2014).

    PubMed  Google Scholar 

  47. Groenendijk, F. H. et al. Sorafenib synergizes with metformin in NSCLC through AMPK pathway activation. Int. J. Cancer 136, 1434–1444 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Ducommun, S. et al. Enhanced activation of cellular AMPK by dual-small molecule treatment: AICAR and A769662. Am. J. Physiol. Endocrinol. Metab. 306, E688–E696 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rohm, M. et al. An AMP-activated protein kinase-stabilizing peptide ameliorates adipose tissue wasting in cancer cachexia in mice. Nat. Med. 22, 1120–1130 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Chen, B. B. et al. Skp-cullin-F box E3 ligase component FBXL2 ubiquitinates Aurora B to inhibit tumorigenesis. Cell Death Dis. 4, e759 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, B. B. et al. F-box protein FBXL2 targets cyclin D2 for ubiquitination and degradation to inhibit leukemic cell proliferation. Blood 119, 3132–3141 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liu, Y. et al. The proapoptotic F-box protein Fbxl7 regulates mitochondrial function by mediating the ubiquitylation and proteasomal degradation of survivin. J. Biol. Chem. 290, 11843–11852 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hebert, A. S. et al. Improved precursor characterization for data-dependent mass spectrometry. Anal. Chem. 90, 2333–2340 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Jurczak, M. J. et al. Dissociation of inositol-requiring enzyme (IRE1α)-mediated c-Jun N-terminal kinase activation from hepatic insulin resistance in conditional X-box-binding protein-1 (XBP1) knock-out mice. J. Biol. Chem. 287, 2558–2567 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported in part by grant no. 16SDG27650008 from the American Heart Association to Y.L. and a US National Institute of Diabetes and Digestive and Kidney Diseases grant to Y.L. and M.J.J. (1R01DK119627). This work was also supported by NHLBI grants nos. UH3HL123502, R01HL096376, R01HL097376, R01HL098174, R01HL081784 and P01HL114453 to R.K.M.; T32HL110849 to T.B.L.; 5R01HL142777 to Y.L.; 5R35HL139860 and 5R01HL133184 to B.B.C. This work was also supported in part by the United States Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development, Biomedical Laboratory Research and Development, a Merit Review Award from the United States Department of Veterans Affairs to R.K.M.

Author information

Authors and Affiliations

Authors

Contributions

Y.L., M.J.J. and B.B.C. designed the study, performed experiments, analyzed results and wrote the manuscript. T.B.L., B.L., M.B.L., J.R.K., M.K.N., F.T., Y.C. and B.R.H. performed experiments and analyzed data. Y.J. designed experiments. S.P.M. directed the NASH human study. C.P.O. directed animal studies. C.P.O., T.F. and R.K.M. edited the manuscript. Y.L., R.K.M. and B.B.C. directed the study.

Corresponding authors

Correspondence to Yuan Liu, Bill B. Chen or Rama K. Mallampalli.

Ethics declarations

Competing interests

A provisional patent application (US patent 62/404,592) has been converted to a Patent Cooperation Treaty application jointly by the US Department of Veterans Affairs and the University of Pittsburgh covering all chemical entities included in this study. Y.L., T.F. and B.B.C. are the founders and employees of Generian Pharmaceuticals and may own company stock.

Additional information

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

Extended data

Extended Data Fig. 1 pAmpkα undergoes ubiquitin proteasomal degradation.

a, BEAS-2B cells were pretreated in glucose free media with increasing concentrations of metformin for 4h. Whole cell extracts were subjected to TUBE agarose beads pull downs (PD) and immunoblotting (Results are representative of three independent experiments). b, Fed and overnight starved mouse liver extracts were subjected to TUBE agarose beads pull down (PD) and immunoblotting (Results are representative of three independent experiments). c, BEAS-2B cells were transfected with either WT or T172A mutant Ampk. Whole cell extracts were subjected to TUBE agarose beads pull down (PD) and immunoblotting. Arrows indicate polyubiquitylated products in each blot (Results are representative of three independent experiments).

Source data

Extended Data Fig. 2 pAmpkα ubiquitylation and degradation is mediated by an F-box protein.

a, BEAS-2B cells were incubated in glucose free 2% DMEM media containing DMSO, MG132 (20 µM, MG) or MLN4924 (5 µM) for 1.5 h. Cells were then collected for immunoblotting (Results are representative of two independent experiments). b, BEAS-2B cells were preincubated in glucose free 2% DMEM media containing DMSO, MG132 (20 µM) or MLN4924 (5 µM) for 1 h. CHX (40 µg/ml) was then added at indicated time points. Cells were collected at same time for immunoblotting (Results are representative of two independent experiments). c, BEAS-2B cells were incubated in high glucose then switched to glucose free 2% DMEM media for 16 h. Total RNA were purified using Qiagen RNeasy Plus Mini Kit. RNA sequencing was then performed by the Quick Biology commercial service. Differentially expressed genes of F-box protein families were plotted and 30% expression level difference was indicated in the red line cut off (Data are the mean of two biological replicates).

Source data

Extended Data Fig. 3 Screening of F-box proteins involved in pAmpkα degradation.

a, BEAS-2B cells were nucleofected with a series of V5 tagged F-box protein encoding plasmids (3 µg). After incubation for 48 h, high glucose media was switched to glucose free 2% FBS DMEM media for 1 h. Cells were then collected for immunoblotting (Results are representative of two independent experiments). b, c, BEAS-2B cells were nucleofected with increasing amounts of either Skp2-V5 or Fbxl5-V5 plasmids. After incubation for 48 h, growth media were changed to glucose free media for 1 h before immunoblotting (Results are representative of two independent experiments). NS indicates a non-specific band. d,. In vitro ubiquitinylation of Ampk. Recombinant ubiquitin E1, E2 conjugating enzymes and ubiquitin was incubated with or without Fbxo48 with pAmpk and polyubiquitinylated products were detected on immunoblots (arrows) (Results are representative of two independent experiments).

Source data

Extended Data Fig. 4 BC1618 off-targeting kinase screen.

The inhibitory activities for BC1618 (10 µM) against 51 kinases (ABL, CSK, EGFR, EPHA2, EPHB4, FGFR1, FLT3, IGF1R, ITK, JAK3, KDR, LCK, MET, PDGFRα, PYK2, SRC, SYK, TIE2, TRKA, TYRO3, AKT1, AMPKα1/β1/γ1, AMPKα2/β1/γ1, AurA, CaMK4, CDK2/CycA2, CHK1, CK1ε, DAPK1, DYRK1B, Erk2, GSK3β, HGK, IKKβ, IRAK4, JNK2, MAPKAPK2, MST1, NEK2, p38α, p70S6K, PAK2, PBK, PDK1, PIM1, PKACα, PKCα, PKD2, ROCK1, SGK, TSSK1) were measured and percentage inhibition of kinase activity was plotted. 50% change of kinase activities is indicated by a red line cut off line (CarnaBio) (Data are the mean of two biological replicates).

Source data

Extended Data Fig. 5 PTM-SEA analysis of BC1618 phosphoproteome.

a, BEAS-2B cells were treated with BC1618 (3µM) or vehicle control and the alterations in the phosphoproteome were assessed using mass spectrometry. Heatmap of those phosphosites with p<0.05 (t-test) comparing BC1618 (left side) to DMSO control (right). Color indicates log2 normalized intensity values as row z-score. Row names indicate the corresponding protein and phosphorylation site. b, Volcano plot of statistical significance (-log10 p. value, y-axis) and the effect size (log2 scaled fold-change, x-axis) of BC1618 treated cells compared to DMSO control, where positive values represent phosphosites higher in BC1618 treated cells. c, Volcano plot of enrichment of various phosphoproteome signatures. The x axis indicates the normalized enrichment score (NES) between BC1618 and DMSO control, with positive values indicating shared phosphosite signatures and negative values indicating opposed phosphosite signatures. Dot size indicates the percent overlap of phosphosites associated with the signature and experimentally assayed phosphosites. Red indicates significantly upregulated signatures while green indicates significantly downregulated signatures (permutation-based FDR < 0.05). Grey indicates non-significant signatures (Data are from three biological replicates).

Source data

Extended Data Fig. 6 BC1618 in vivo toxicity studies.

(a-d) C57BL/6 mice were given BC1618 in drinking water at 15 mg/kg/d (low) and 30 mg/kg/d (high) doses for 3 months. Mice were then euthanized, and plasma samples were collected and assayed for markers of cytotoxicity. Alanine aminotransferase (ALT) activity, creatine kinase activity, lactate dehydrogenase (LDH) content, and creatinine content were measured following the manufacturer’s protocol. e, Major organs were collected and processed for H&E staining; scale bar: 100 μm. Data represented mean +/- SEM. Data points represent n=7–8 independent mice per group, and p-values are indicated, as calculated by one-way ANOVA with Dunnett’s test of multiple comparisons (A-D).

Source data

Extended Data Fig. 7 BC1618 is anti-inflammatory in vitro.

a, THP1 cells stably expressing an NF-κB inducible luciferase (secreted) reporter were treated with LPS (100 ng/ml) and BC1618 at indicated concentrations for 6 h or 24 h before supernatant was collected for luciferase activity assay following the manufacturer’s protocol. Data are shown as the mean ± SEM of three independent biological replicates, and significance was measured by one-way ANOVA with Dunnett’s test of multiple comparisons relative to 0µM BC1618+LPS. For 6hs, 0.0032µM: p=0.0868; for 0.016,0.08, 0.4, 2, and 10µM: p<0.0001. For 24hr, 0.0032µM: p=0.0002; for 0.016,0.08, 0.4, 2, and 10µM: p<0.0001. b, 50K PBMC cells were cultured in 96 well plates before being exposed to BC1618 at indicated concentrations for 18 h. Cells were then treated with LPS (10 ng/ml) for an additional 4 h. Media were then collected and TNF and IL-1 concentration were determined by ELISA. Data are shown as the mean ± SEM of three independent biological replicates, and significance was measured by one-way ANOVA with Dunnett’s test of multiple comparisons relative to 0µM BC1618 for both cytokines. For TNF treatment, 0.008µM: p=0.2212; 0.04µM: p=0.0375; 0.2µM: p=0.3585; for 1, 5, and 25µM: p<0.0001. For IL-1 treatment, 0.008µM: p=0.7497; 0.04µM: p=0.0919; 0.2µM: p=0.0126; 1µM: p=0.0028; 5µM: p=0.0015; 25µM: p=0.0065. Significance is also indicated as follows: *, P <0.05; **, P < 0.01; ***, P <0.001; ****, P<0.0001 (c) PBMC cells (1 mL at 1.0 x 106/ml) were treated with BC1618 (5 µM) for 18 h. Cells were then treated with 100 ng/ml LPS for additional 4 h. Cytokine release was monitored by the human cytokine array (R&D systems). The results from a cytokine array dot blot are shown and quantitated (Data are shown as the mean of two independent biological replicates).

Source data

Extended Data Fig. 8 BC1618 reduces lung inflammation in endotoxin treated mice.

C57BL/6 mice were administered i.p. with vehicle, 2 or 10 mg/kg of BC1618. Mice were then immediately challenged with LPS (3 mg/kg) for an additional 18 h. Mice were euthanized and lungs were lavaged with saline, harvested, and then homogenized. Bronchoalveolar lavage (BAL) protein (a), cell counts (b) and cytokines (c-e) were measured. f, Representative mouse lung tissue H&E staining; scale bar: 100 μm. Data represented mean +/- SEM (n=5 independent mice per group). P-values are indicated as calculated by one-way ANOVA with Dunnett’s test of multiple comparisons (A-E).

Source data

Extended Data Fig. 9 BC1618 increases Ampkα protein levels in mice after dietary restriction.

a, b, C57BL/6 mice were intraperitoneally (i.p.) administered with vehicle or BC1618 (20 mg/kg) on day 1 and simultaneously chow was removed for overnight starvation. After a 17 h fast, mice were given vehicle or BC1618 i.p. 30 min later, mice were euthanized, PBS was perfused through heart to remove blood. Heart, liver, and skeletal muscle was collected and fresh frozen for tissue homogenization and immunoblotting. The pAmpkα and Ampkα protein levels were quantitated and plotted in b. Densitometry data were corrected to tubulin and normalized to vehicle. Data represented mean +/- SEM (n=4-5 independent mice per group). P-values are indicated as calculated by two-tailed unpaired t-test (B).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–10, Supplementary Fig. gel source data and Note.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed immunoblots.

Source Data Fig. 2

Unprocessed immunoblots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed immunoblots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed immunoblots.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed immunoblots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed immunoblots.

Source Data Extended Data Fig. 3

Unprocessed immunoblots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed immunoblots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Jurczak, M.J., Lear, T.B. et al. A Fbxo48 inhibitor prevents pAMPKα degradation and ameliorates insulin resistance. Nat Chem Biol 17, 298–306 (2021). https://doi.org/10.1038/s41589-020-00723-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-00723-0

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