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:

Mitochondrial uncouplers induce proton leak by activating AAC and UCP1

Abstract

Mitochondria generate heat due to H+ leak (IH) across their inner membrane1. IH results from the action of long-chain fatty acids on uncoupling protein 1 (UCP1) in brown fat2,3,4,5,6 and ADP/ATP carrier (AAC) in other tissues1,7,8,9, but the underlying mechanism is poorly understood. As evidence of pharmacological activators of IH through UCP1 and AAC is lacking, IH is induced by protonophores such as 2,4-dinitrophenol (DNP) and cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP)10,11. Although protonophores show potential in combating obesity, diabetes and fatty liver in animal models12,13,14, their clinical potential for treating human disease is limited due to indiscriminately increasing H+ conductance across all biological membranes10,11 and adverse side effects15. Here we report the direct measurement of IH induced by DNP, FCCP and other common protonophores and find that it is dependent on AAC and UCP1. Using molecular structures of AAC, we perform a computational analysis to determine the binding sites for protonophores and long-chain fatty acids, and find that they overlap with the putative ADP/ATP-binding site. We also develop a mathematical model that proposes a mechanism of uncoupler-dependent IH through AAC. Thus, common protonophoric uncouplers are synthetic activators of IH through AAC and UCP1, paving the way for the development of new and more specific activators of these two central mediators of mitochondrial bioenergetics.

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: H+ currents activated by DNP and FCCP in the IMM versus plasma membrane.
Fig. 2: DNP and FCCP activate IH through AAC.
Fig. 3: DNP and FCCP induce IH through UCP1.
Fig. 4: Uncouplers and FAs bind within AAC.

Similar content being viewed by others

Data availability

All data and materials are available on request from the corresponding authors.

Code availability

The MATLAB code for the mathematical model, force field and docking parameters for DNP, FCCP, BAM15, SF6847, CATR and the homology model of m-state bovine AAC1 are available at Zenodo (https://doi.org/10.5281/zenodo.5058463).

References

  1. Divakaruni, A. S. & Brand, M. D. The regulation and physiology of mitochondrial proton leak. Physiology 26, 192–205 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Fedorenko, A., Lishko, P. V. & Kirichok, Y. Mechanism of fatty-acid-dependent UCP1 uncoupling in brown fat mitochondria. Cell 151, 400–413 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nicholls, D. G. A history of UCP1. Biochem. Soc. Trans. 29, 751–755 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Aquila, H., Link, T. A. & Klingenberg, M. The uncoupling protein from brown fat mitochondria is related to the mitochondrial ADP/ATP carrier. Analysis of sequence homologies and of folding of the protein in the membrane. EMBO J. 4, 2369–2376 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bouillaud, F., Weissenbach, J. & Ricquier, D. Complete cDNA-derived amino acid sequence of rat brown fat uncoupling protein. J. Biol. Chem. 261, 1487–1490 (1986).

    Article  CAS  PubMed  Google Scholar 

  6. Klingenberg, M. & Huang, S. G. Structure and function of the uncoupling protein from brown adipose tissue. Biochim. Biophys. Acta 1415, 271–296 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Bertholet, A. M. et al. H+ transport is an integral function of the mitochondrial ADP/ATP carrier. Nature 571, 515–520 (2019).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  8. Andreyev, A. et al. The ATP/ADP-antiporter is involved in the uncoupling effect of fatty acids on mitochondria. Eur. J. Biochem. 182, 585–592 (1989).

    Article  PubMed  Google Scholar 

  9. Brustovetsky, N. & Klingenberg, M. The reconstituted ADP/ATP carrier can mediate H+ transport by free fatty acids, which is further stimulated by mersalyl. J. Biol. Chem. 269, 27329–27336 (1994).

    Article  CAS  PubMed  Google Scholar 

  10. McLaughlin, S. G. & Dilger, J. P. Transport of protons across membranes by weak acids. Physiol. Rev. 60, 825–863 (1980).

    Article  CAS  PubMed  Google Scholar 

  11. Terada, H. Uncouplers of oxidative phosphorylation. Environ. Health Perspect. 87, 213–218 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Perry, R. J., Zhang, D., Zhang, X. M., Boyer, J. L. & Shulman, G. I. Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats. Science 347, 1253–1256 (2015).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  13. Perry, R. J. et al. Reversal of hypertriglyceridemia, fatty liver disease, and insulin resistance by a liver-targeted mitochondrial uncoupler. Cell Metab. 18, 740–748 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tainter, M. L., Stockton, A. B. & Cutting, W. C. Use of dinitrophenol in obesity and related conditions. J. Am. Med. Assoc. 101, 1472–1475 (1933).

    Article  Google Scholar 

  15. Grundlingh, J., Dargan, P. I., El-Zanfaly, M. & Wood, D. M. 2,4-Dinitrophenol (DNP): a weight loss agent with significant acute toxicity and risk of death. J. Med. Toxicol. 7, 205–212 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lowell, B. B. & Spiegelman, B. M. Towards a molecular understanding of adaptive thermogenesis. Nature 404, 652–660 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Krauss, S., Zhang, C. Y. & Lowell, B. B. The mitochondrial uncoupling-protein homologues. Nat. Rev. Mol. Cell Biol. 6, 248–261 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Skulachev, V. P. Uncoupling: new approaches to an old problem of bioenergetics. Biochim. Biophys. Acta 1363, 100–124 (1998).

    Article  CAS  PubMed  Google Scholar 

  19. Bertholet, A. M. et al. Mitochondrial patch clamp of beige adipocytes reveals UCP1-positive and UCP1-negative cells both exhibiting futile creatine cycling. Cell Metab. 25, 811–822 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bertholet, A. M. & Kirichok, Y. Patch-clamp analysis of the mitochondrial H+ leak in brown and beige fat. Front. Physiol. 11, 326 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Levy, S. E., Chen, Y. S., Graham, B. H. & Wallace, D. C. Expression and sequence analysis of the mouse adenine nucleotide translocase 1 and 2 genes. Gene 254, 57–66 (2000).

    Article  CAS  PubMed  Google Scholar 

  22. Rodic, N. et al. DNA methylation is required for silencing of ant4, an adenine nucleotide translocase selectively expressed in mouse embryonic stem cells and germ cells. Stem Cells 23, 1314–1323 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Cho, J. et al. Mitochondrial ATP transporter Ant2 depletion impairs erythropoiesis and B lymphopoiesis. Cell Death Differ. 22, 1437–1450 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Graham, B. H. et al. A mouse model for mitochondrial myopathy and cardiomyopathy resulting from a deficiency in the heart/muscle isoform of the adenine nucleotide translocator. Nat. Genet. 16, 226–234 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Childress, E. S., Alexopoulos, S. J., Hoehn, K. L. & Santos, W. L. Small molecule mitochondrial uncouplers and their therapeutic potential. J. Med. Chem. 61, 4641–4655 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Kenwood, B. M. et al. Identification of a novel mitochondrial uncoupler that does not depolarize the plasma membrane. Mol. Metab. 3, 114–123 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Bernardi, P. Mitochondrial transport of cations: channels, exchangers, and permeability transition. Physiol. Rev. 79, 1127–1155 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Klingenberg, M. The ADP and ATP transport in mitochondria and its carrier. Biochim. Biophys. Acta 1778, 1978–2021 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Alexopoulos, S. J. et al. Mitochondrial uncoupler BAM15 reverses diet-induced obesity and insulin resistance in mice. Nat. Commun. 11, 2397 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  30. Axelrod, C. L. et al. BAM15-mediated mitochondrial uncoupling protects against obesity and improves glycemic control. EMBO Mol. Med. 12, e12088 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Cannon, B. & Nedergaard, J. Brown adipose tissue: function and physiological significance. Physiol. Rev. 84, 277–359 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Bertholet, A. M. & Kirichok, Y. UCP1: a transporter for H+ and fatty acid anions. Biochimie 134, 28–34 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Nicholls, D. G. & Lindberg, O. Brown-adipose-tissue mitochondria. The influence of albumin and nucleotides on passive ion permeabilities. Eur. J. Biochem. 37, 523–530 (1973).

    Article  CAS  PubMed  Google Scholar 

  34. Klingenberg, M. UCP1—a sophisticated energy valve. Biochimie 134, 19–27 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Pebay-Peyroula, E. et al. Structure of mitochondrial ADP/ATP carrier in complex with carboxyatractyloside. Nature 426, 39–44 (2003).

    Article  CAS  PubMed  ADS  Google Scholar 

  36. Nury, H. et al. Structural basis for lipid-mediated interactions between mitochondrial ADP/ATP carrier monomers. FEBS Lett. 579, 6031–6036 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Ruprecht, J. J. et al. Structures of yeast mitochondrial ADP/ATP carriers support a domain-based alternating-access transport mechanism. Proc. Natl Acad. Sci. USA 111, E426–E434 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kunji, E. R. & Robinson, A. J. The conserved substrate binding site of mitochondrial carriers. Biochim. Biophys. Acta 1757, 1237–1248 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Dehez, F., Pebay-Peyroula, E. & Chipot, C. Binding of ADP in the mitochondrial ADP/ATP carrier is driven by an electrostatic funnel. J. Am. Chem. Soc. 130, 12725–12733 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Ruprecht, J. J. et al. The molecular mechanism of transport by the mitochondrial ADP/ATP carrier. Cell 176, 435–447 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bielawski, J., Thompson, T. E. & Lehninger, A. L. The effect of 2,4-dinitrophenol on the electrical resistance of phospholipid bilayer membranes. Biochem. Biophys. Res. Commun. 24, 948–954 (1966).

    Article  CAS  PubMed  Google Scholar 

  42. Skulachev, V. P., Sharaf, A. A. & Liberman, E. A. Proton conductors in the respiratory chain and artificial membranes. Nature 216, 718–719 (1967).

    Article  CAS  PubMed  ADS  Google Scholar 

  43. Skulachev, V. P. et al. The effect of uncouplers on mitochondria, respiratory enzyme complexes and artificial phospholipid membranes. Curr. Mod. Biol. 2, 98–105 (1968).

    CAS  PubMed  Google Scholar 

  44. Mitchell, P. & Moyle, J. Stoichiometry of proton translocation through the respiratory chain and adenosine triphosphatase systems of rat liver mitochondria. Nature 208, 147–151 (1965).

    Article  CAS  PubMed  ADS  Google Scholar 

  45. Mitchell, P. Chemiosmotic coupling in oxidative and photosynthetic phosphorylation. Biol. Rev. Camb. Philos. Soc. 41, 445–502 (1966).

    Article  CAS  PubMed  Google Scholar 

  46. Betz, M. J. & Enerback, S. Human brown adipose tissue: what we have learned so far. Diabetes 64, 2352–2360 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Reynafarje, B., Costa, L. E. & Lehninger, A. L. O2 solubility in aqueous media determined by a kinetic method. Anal. Biochem. 145, 406–418 (1985).

    Article  CAS  PubMed  Google Scholar 

  48. Olsson, M. H., Sondergaard, C. R., Rostkowski, M. & Jensen, J. H. PROPKA3: consistent treatment of internal and surface residues in empirical pKa predictions. J. Chem. Theory Comput. 7, 525–537 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Grimme, S., Antony, J., Ehrlich, S. & Krieg, H. A consistent and accurate ab initio parametrization of density functional dispersion correction (DFT-D) for the 94 elements H-Pu. J. Chem. Phys. 132, 154104 (2010).

    Article  PubMed  ADS  CAS  Google Scholar 

  50. Roos, K. et al. OPLS3e: extending force field coverage for drug-like small molecules. J. Chem. Theory Comput. 15, 1863–1874 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. Friesner, R. A. et al. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J. Med. Chem. 49, 6177–6196 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. 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 

  53. Jo, S., Kim, T. & Im, W. Automated builder and database of protein/membrane complexes for molecular dynamics simulations. PLoS ONE 2, e880 (2007).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  54. Comte, J., Maisterrena, B. & Gautheron, D. C. Lipid composition and protein profiles of outer and inner membranes from pig heart mitochondria. Comparison with microsomes. Biochim. Biophys. Acta 419, 271–284 (1976).

    Article  CAS  PubMed  Google Scholar 

  55. Huang, J. et al. CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14, 71–73 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. Klauda, J. B. et al. Update of the CHARMM all-atom additive force field for lipids: validation on six lipid types. J. Phys. Chem. B 114, 7830–7843 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Jorgensen, W. L., Chandrasekhar, J., Madura, J. D., Impey, R. W. & Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 79, 926–935 (1983).

    Article  CAS  ADS  Google Scholar 

  58. Beglov, D. & Roux, B. Finite representation of an infinite bulk system: solvent boundary potential for computer simulations. J. Chem. Phys. 100, 9050–9063 (1994).

    Article  CAS  ADS  Google Scholar 

  59. Vanommeslaeghe, K. et al. CHARMM general force field: a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J. Comput. Chem. 31, 671–690 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Mayne, C. G., Saam, J., Schulten, K., Tajkhorshid, E. & Gumbart, J. C. Rapid parameterization of small molecules using the Force Field Toolkit. J. Comput. Chem. 34, 2757–2770 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Frisch, M. J. et al. Gaussian 16 rev. C.01 (Gaussian, 2016).

  62. Abraham, M. J. et al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 1–2, 19–25 (2015).

    Article  ADS  Google Scholar 

  63. Hoover, W. G. Canonical dynamics: equilibrium phase-space distributions. Phys. Rev. A Gen. Phys. 31, 1695–1697 (1985).

    Article  CAS  PubMed  ADS  Google Scholar 

  64. Nosé, S. A molecular dynamics method for simulations in the canonical ensemble. Mol. Phys. 52, 255–268 (1984).

    Article  ADS  Google Scholar 

  65. Bussi, G., Donadio, D. & Parrinello, M. Canonical sampling through velocity rescaling. J. Chem. Phys. 126, 014101 (2007).

    Article  PubMed  ADS  CAS  Google Scholar 

  66. Parrinello, M. & Rahman, A. Polymorphic transitions in single crystals: a new molecular dynamics method. J. Appl. Phys. 52, 7182–7190 (1981).

    Article  CAS  ADS  Google Scholar 

  67. Darden, T., York, D. & Pedersen, L. Particle mesh Ewald: an Nlog(N) method for Ewald sums in large systems. J. Chem. Phys. 98, 10089–10092 (1993).

    Article  CAS  ADS  Google Scholar 

  68. Hess, B., Bekker, H., Berendsen, H. J. C. & Fraaije, J. G. E. M. LINCS. J. Comput. Chem. 18, 1463–1472 (1997).

    Article  CAS  Google Scholar 

  69. Gumbart, J., Khalili-Araghi, F., Sotomayor, M. & Roux, B. Constant electric field simulations of the membrane potential illustrated with simple systems. Biochim. Biophys. Acta 1818, 294–302 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. Michaud-Agrawal, N., Denning, E. J., Woolf, T. B. & Beckstein, O. MDAnalysis: a toolkit for the analysis of molecular dynamics simulations. J. Comput. Chem. 32, 2319–2327 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996). 27-38.

    Article  CAS  PubMed  Google Scholar 

  72. Marcoline, F. V., Bethel, N., Guerriero, C. J., Brodsky, J. L. & Grabe, M. Membrane protein properties revealed through data-rich electrostatics calculations. Structure 23, 1526–1537 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Baker, N. A., Sept, D., Joseph, S., Holst, M. J. & McCammon, J. A. Electrostatics of nanosystems: application to microtubules and the ribosome. Proc. Natl Acad. Sci. USA 98, 10037–10041 (2001).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

Download references

Acknowledgements

We thank D. C. Wallace for providing AAC1-knockout mice; A. Angelin for performing pilot mitochondrial respiration studies; and B. M. Spiegelman for the help with Seahorse respiration studies. This work was supported by NIH grants R01GM107710, R01GM118939 and R35GM136415 to Y.K., R01GM089740 and R01GM137109 to M.G., R01NS098772 to N.B., as well as a grant from the UCSF Program for Breakthrough Biomedical Research (PBBR) to M.G. and Y.K. A.M.B. was supported by an American Heart Association Career Development Award 19CDA34630062 and NIH grant R35GM143097. P.B. was supported by an American Heart Association Post-Doctoral Fellowship 18POST33960587. Simulations were carried out, in part, at the UCSF Wynton Cluster on hardware made possible through NIH grants 1S10OD021596 and R01GM089740 to M.G.

Author information

Authors and Affiliations

Authors

Contributions

A.M.B., A.M.N., P.B., M.G. and Y.K. conceived the project and designed experiments. A.M.B. performed all of the electrophysiological experiments, except for pilot experiments and data for Fig. 1, which were performed by A.F.; A.M.B, E.T.C., L.K., J.H., T.B. and N.B. performed respirometry on C2C12 mitochondria. J.S. performed respiration experiments in C2C12 cells. A.M.N., P.B. and M.G. designed and performed computational analyses. A.M.B., A.M.N., P.B., M.G. and Y.K. wrote the manuscript. All of the authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Michael Grabe or Yuriy Kirichok.

Ethics declarations

Competing interests

Y.K., M.G., P.B. and J.S. are shareholders of Equator Therapeutics. Y.K. is an advisor to Equator Therapeutics. Y.K. is shareholder of YourChoice Therapeutics. E.T.C. is a founder, board member and equity holder in Matchpoint Therapeutics. The other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Oliver Beckstein, Danica Chen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 DNP- and FCCP-induced mitochondrial IH.

a, IH densities induced by different DNP concentrations in heart IMM at −160 mV (n = 15 for 50 µM, n = 9 for 100 µM, n = 8 for 200 µM, n = 14 for 500 µM). Data are mean ± SEM. b, IH induced by 50 μM DNP in heart IMM before and after application 4 μM BKA (left). Remaining fraction of IH induced by 50 μM DNP at −160 mV after inhibition by 4 μM BKA, n = 3 (right). Data are mean ± SEM. c, IH induced by 500 μM DNP on the background of 10 mM MβCD in heart IMM before and after application of 1 µM CATR (left). IH densities induced by 50 µM DNP alone (n = 15) and 500 µM DNP on the background of 10 mM MβCD (n = 6) (right). Data are mean ± SEM. d, IH induced by 250 nM FCCP in heart IMM was inhibited by 1 μM CATR. e, Remaining fraction of FCCP-induced IH at -160 mV after inhibition by 1 μM CATR. Control (100% IH) is shown in gray. Data are mean ± SEM. Paired t-test, two-tailed, control vs. 1 µM CATR, n = 6.

Extended Data Fig. 2 SF6847 and BAM15 induce IH via AAC.

a, IH induced by 25 nM SF6847 across WT heart IMM before and after application 1 μM CATR. b, IH induced by 25 nM SF6847 in AAC1−/− heart IMM. c, Remaining fraction of IH induced by 25 nM SF6847 at −160 mV after inhibition by 1 μM CATR. Data are mean ± SEM. Paired t-test, two-tailed, control vs CATR, n = 6. d, IH densities induced by 25 nM SF6847 at −160 mV in WT (n = 10) and AAC1−/− (n = 5) heart IMM. IH was measured at −160 mV. Data are mean ± SEM. Mann–Whitney U-test, two-tailed, WT vs AAC1−/−. e, IH induced by 250 nM BAM15 across WT heart IMM before and after application 1 μM CATR. f, IH induced by 250 nM BAM15 in AAC1−/− heart IMM. g, Remaining fraction of IH induced by 250 nM BAM15 at −160 mV after inhibition by 1 μM CATR. Data are mean ± SEM. Paired t-test, two-tailed, control vs CATR, n = 4. h, IH densities induced by 250 nM BAM15 at −160 mV in WT (n = 5) and AAC1−/− (n = 5) heart IMM. IH was measured at −160 mV. Data are mean ± SEM. Mann–Whitney U-test, two-tailed, WT vs AAC1−/−. i, j, Representative K+ currents induced by 100 nM valinomycin across heart IMM in WT (i) and AAC1−/− (j). k, K+ current densities induced by 100 nM valinomycin at −160 mV in WT and AAC1−/− heart IMM (n = 5). Data are mean ± SEM.

Extended Data Fig. 3 DNP- and BAM-induced mitochondrial uncoupled respiration require AAC.

a, Oxygen consumption rate (OCR) of mitochondria isolated from WT and DKO C2C12 cells in the presence of oligomycin as measured with the Seahorse Analyzer. Each point corresponds to an individual respiration well, n = –60 (WT), and n = 56 (DKO). Mann–Whitney U-test, two-tailed. Data represent mean ± SEM. b, c OCR increase observed in mitochondria isolated from WT and DKO C2C12 cells after addition of DNP, n = 20, WT and n = 17, DKO (b) or BAM15, n = 10, WT and n = 10, DKO (c) in the presence of oligomycin. Each point corresponds to OCR change in an individual well of the Seahorse Analyzer, n = 17 – 20 (DNP) and n = 10 (BAM15). Data are mean ± SEM. Mann–Whitney U-test, two-tailed. d, OCR time course of isolated mitochondria from WT (n = 20 wells, blue) and DKO C2C12 cells (n = 17 wells, red). Basal respiration (1), addition of oligomycin (2), addition of DNP (3) and addition of rotenone/antimycin (4). Data represent mean ± SEM. e, Basal OCR of mitochondria isolated from WT or DKO C2C12 cells measured with a Clark electrode (n = 22, WT and n = 14, DKO). Mann–Whitney U-test, two-tailed. Data represent mean ± SEM. f, OCR increase above basal, observed using a Clark electrode after addition of DNP to mitochondria isolated from WT and DKO C2C12 cells; n = 3 (WT) and n = 5 (DKO) for 5 µM; n = 5 (WT) and n = 4 (DKO) for 12.5 µM. Data are mean ± SEM. Mann–Whitney U-test, two-tailed. g, Left panel: Immunoblots of WT (n = 5) and DKO (n = 5) C2C12 cells showing expression of the electron-transport chain complexes complex I (CI) – complex V (CV), TOM20, and the loading control (plasma membrane Na+/K+ ATPase). Right panel: The same immunoblots at longer exposure for the bands corresponding to complexes I-V. h, TOM20 protein expression level relative to Na+/K+ ATPase for the immunoblot shown in (g). Data shown as mean ± SEM, n = 5. Two-tailed unpaired t-test, WT vs DKO. im, Protein expression levels for complexes I-V relative to TOM20 for the immunoblots shown in (g). Data shown as mean ± SEM, n = 5. Two-tailed unpaired t-test, WT vs DKO. For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 4 Respiration induced by mitochondrial uncouplers in intact cells depends on AAC.

a, Oxygen consumption rate (OCR) of WT (n = 20) and DKO C2C12 cells (n = 16) before and after addition of oligomycin (oligo). Two-tailed unpaired t-test, WT vs DKO. bd, OCR increase observed in WT and DKO C2C12 cells upon application of various concentrations of FCCP (n = 4), DNP (n = 4), and BAM15 (n = 4). All OCRs were measured in the presence of oligomycin. Two-tailed unpaired t-test, WT vs DKO. e, AAC-dependent fraction of uncoupled respiration at various concentrations of FCCP, DNP, and BAM15. All data shown as mean ± SEM, n = 4. f, OCR increase observed in WT and DKO C2C12 cells upon application of various concentrations of valinomycin. All OCRs were measured in the presence of oligomycin. Nigericin (1 µM) was added to reduce matrix K+ accumulation and mitochondrial swelling caused by valinomycin27. Two-tailed unpaired t-test, WT vs DKO, n = 4.

Extended Data Fig. 5 Adenine nucleotides inhibit DNP-induced IH.

a, DNP-induced IH at various ATP concentrations on the cytosolic face of the IMM of brown fat. IH was activated with either 500 μM DNP (left panel) or 5 mM DNP (middle panel) applied in the presence of 10 mM MβCD. Control currents were recorded in 10 mM MβCD. Right panel is the dose-dependence of IH inhibition by ATP at 500 μM DNP (red, n = 5) and 5 mM DNP (blue, n = 6). Current amplitudes were measured at −160 mV. Data are mean ± SEM. b, Left panel, IH induced by 50 µM DNP in control (1), upon transient inhibition by bath ADP (2), and upon subsequent recovery in the presence ADP (3). Heart mitoplast. Middle panel, DNP-dependent IH time course of left panel. IH was measured at −160 mV. Right panel, ADP inhibition of IH at points (2) and (3) in left and middle panels. Data are mean ± SEM. Paired t-test, two-tailed, ADP (2) vs. control, n = 4. c, IH activated by 50 µM DNP was inhibited by bath ADP. Pipette solution contained 1 mM ADP. Heart mitoplast. Middle panel, IH time course of the left panel. Right panel, ADP inhibition of IH in point (2) as in left and middle panels. Data are mean ± SEM. Paired t-test, two-tailed, n = 4.

Extended Data Fig. 6 Binding mode comparisons of CATR, ADP, small molecule uncouplers, and fatty acid.

ad, Comparison of binding poses in the AAC1 c-state (2C3E) cavity of (a) docked ADP and docked DNP; (b) crystallographic CATR and docked DNP; (c) FA (arachidonic acid) pose from MD simulation and docked ADP; and (d) FA pose from MD simulation and crystallographic CATR. TM helices 1 and 6 of AAC1 are hidden. ADP and CATR are shown as thinner sticks with mauve carbon atoms. DNP and FA are shown as thicker sticks with yellow carbon atoms. e, Full final docked DNP, FCCP, BAM15, and SF6847 poses in their deprotonated and protonated forms (left) as in Fig. 4c and titratable heavy atoms (pink spheres) from these poses (right). TM helix 1 of AAC1 is hidden. f, ADP binding mode to the c-state as predicted from docking. g, Simulation snapshot showing arachidonic acid (solid spheres) bound to the TM5/6 fenestration of AAC1 in the c-state superimposed with the docked pose of DNP (transparent spheres). h, Simulation snapshot showing two arachidonic acid molecules bound simultaneously to the AAC1 c-state cavity, one in the TM5/6 fenestration and the other in the small molecule uncoupler site.

Extended Data Fig. 7 Docking reveals a common binding site for small molecules and nucleotide in both c- and m-states of AAC1.

aj, Final docking poses to the AAC1 c-state (ae) and m-state (fj) structures of deprotonated forms of DNP, FCCP, BAM15, and SF6847, as well as ADP, respectively. In e and j ADP phosphate groups are hidden for clarity but were included in the docking. The same protein sidechains are shown as sticks in all panels.

Extended Data Fig. 8 DNP binds to the c-state of AAC1 in MD simulations.

a, Illustration of the simulation setup used to assess binding of negatively charged DNP to the c-state of AAC1. DNP is shown in sphere representation in the aqueous region of the simulation box. AAC1 is shown as a cyan ribbon, with TM1 and TM6 hidden, and lipid molecules are shown as sticks. The Cζ atom of AAC1 residue R234 at the base of the c-state cavity is shown as a black sphere. Binding was tracked in bd by monitoring the distance from the DNP centre of mass to AAC1 R234 Cζ atom indicated by the dotted line. b, Trajectories of negatively charged DNP initially placed in solution far from the binding site. c, Trajectories of neutral DNP, initially placed in the binding site. d, Trajectories of negatively charged DNP initially in the binding site, with an applied −160 mV membrane potential. In each plot, the two different colored traces are measurements made from two independent simulation trajectories. Simulations in c were initiated from docking poses, while those under an applied −160 mV potential in d were initiated from the final snapshots of the DNP binding simulations in b.

Extended Data Fig. 9 Fatty acids bind to the c-state of AAC1 via a TM5/6 fenestration.

a, Arachidonic acid transiently bound to the AAC1 region identified as the DNP/small molecule binding site in contact with protein residue Y186; snapshots are from two independent simulation trajectories. View is from the membrane with TM5 & TM6 of AAC1 hidden to show the cavity. b, Top view of structures in panel a viewed from the cytoplasm. c, Final states of four arachidonic and three palmitic acids bound to the fenestration between AAC1 helices TM5 & TM6; snapshots are from 7 independent simulation trajectories. d, Top view of structures in panel c. In panels ad, AAC1 is shown as a cyan ribbon and FAs are shown as sticks with yellow carbon and red oxygen atoms. e, Side view and f, top view of a single structure from panels c and d with FA atoms shown as spheres and bilayer lipids shown as sticks. Carbon atoms 1–6, 7–12, and 13–20 of the arachidonic acid are colored yellow, blue, and pink, to highlight the parts that are inside the AAC1 cavity, in the TM5/TM6 fenestration, and interacting with bilayer lipids, respectively.

Extended Data Fig. 10 Calculation of membrane potential profiles and mathematical model results for AAC1.

a, Membrane potential profile (V(z)) across c-state (left) and energy values along the central pore for a monovalent cation (|e|·V(z)) (right). The profiles were computed under a −160 mV applied voltage using Poisson-Boltzmann theory (see Materials and Methods) on a configuration extracted from the equilibrated MD simulations. Colour gradients represent isocontours of linearly spaced scalar values ranging from 0 mV (upper region of graphic) to −160 mV (lower region). The water filled cavity facing upward supports a minor fraction of the field with the majority of the field focused across the closed matrix gate of the c-state. These values are quantified in the right panel showing the energy of interaction for a monovalent cation moving through the membrane field. be, Steady state single-transporter currents as a function of voltage (b, c), pKa (d), and rate k23 of proton transfer from the uncoupler to matrix pathway (e) for the 3-state model shown in Fig. 4g using values in Supplementary Table 2 unless otherwise indicated in the panel. The electrostatic potential in the cavity (Eelec) was varied: 10 kBT (blue), 8 kBT (gold), 6 kBT (green) (b and d). The fraction of the membrane voltage traversed by a H+ entering from the cytosol was varied: f = 0.1 (blue), 0.25 (gold), and 0.5 (green) (c). The energy of the proton in the matrix pathway (E1) was varied: 0 kBT (blue), 5 kBT (gold), 10 kBT (green) (e).

Supplementary information

Supplementary Information

Supplementary Fig. 1: gel source data; Supplementary Note 1: mathematical model of uncoupler-aided H+ permeation through AAC; Supplementary Note 2: Supplementary Discussion; Supplementary Table 1: parameters for electrostatic calculations; Supplementary Table 2: base mathematical model parameters; and Supplementary Table 3: molecular dynamics simulations.

Reporting Summary

Supplementary Video 1

DNP binding to AAC1 in two independent molecular dynamics simulations. Two independent 500 ns simulation trajectories of AAC1 (cyan ribbon) with charged DNP (yellow/blue/red sticks), shown side by side. Distances from DNP to the bottom of the AAC1 cavity are plotted in Extended Data Fig. 8b. The transparent, fixed DNP molecule represents the DNP pose identified from docking. White spheres are selected lipid atoms indicating the extent of the hydrophobic interior of the lipid bilayer.

Supplementary Video 2

Arachidonic acid binding to AAC1 in two independent molecular dynamics simulations. Two independent 5 µs simulation trajectories of AAC1 (cyan ribbon) with arachidonic acid (yellow and red spheres), shown side by side. Each shows the arachidonic acid binding first to the DNP/uncoupler site, and then to the fenestration between TM5 and TM6. White spheres are selected lipid atoms indicating the extent of the hydrophobic interior of the lipid bilayer.

Supplementary Video 3

The AAC1 c-state can accommodate two FA molecules simultaneously. A 1 µs simulation of two arachidonic acid molecules (yellow and red spheres) simultaneously occupying the DNP/uncoupler site and the FA site between TM5 and TM6. AAC1 is shown as a cyan ribbon and is viewed from the cytoplasmic side of the membrane. Sidechains of protein residues Lys22, Arg79, Tyr186 and Arg279 are shown as sticks.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bertholet, A.M., Natale, A.M., Bisignano, P. et al. Mitochondrial uncouplers induce proton leak by activating AAC and UCP1. Nature 606, 180–187 (2022). https://doi.org/10.1038/s41586-022-04747-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04747-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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