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:

Fatty acyl-coenzyme A activates mitochondrial division through oligomerization of MiD49 and MiD51

Abstract

Mitochondrial fission occurs in many cellular processes, but the regulation of fission is poorly understood. We show that long-chain acyl-coenzyme A (LCACA) activates two related mitochondrial fission proteins, MiD49 and MiD51, by inducing their oligomerization, which activates their ability to stimulate the DRP1 GTPase. The 1:1 stoichiometry of LCACA:MiD in the oligomer suggests interaction in the previously identified nucleotide-binding pocket, and a point mutation in this pocket reduces LCACA binding and LCACA-induced oligomerization for MiD51. In cells, this LCACA binding mutant does not assemble into puncta on mitochondria or rescue MiD49/51 knockdown effects on mitochondrial length and DRP1 recruitment. Furthermore, cellular treatment with BSA-bound oleic acid, which causes increased LCACA, promotes mitochondrial fission in an MiD49/51-dependent manner. These results suggest that LCACA is an endogenous ligand for MiDs, inducing mitochondrial fission and providing a potential mechanism for fatty-acid-induced mitochondrial division. Finally, MiD49 or MiD51 oligomers synergize with Mff, but not with actin filaments, in DRP1 activation, suggesting distinct pathways for DRP1 activation.

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: Long-chain acyl-CoA induces MiD49 oligomerization.
Fig. 2: LCACA-induced MiD49 oligomers activate DRP1 in a synergistic manner with Mff.
Fig. 3: MiD51 oligomerizes in the presence of long-chain acyl-CoA.
Fig. 4: Mutation of R342 in MiD51 reduces LCACA-induced oligomerization.
Fig. 5: MiD51-LCACA binding mutant does not form mitochondria-associated puncta in cells.
Fig. 6: MiD51-LCACA binding mutant does not rescue mitochondrial elongation caused by MiD49/51 suppression.
Fig. 7: MiD51-LCACA binding mutant does not rescue mitochondrial elongation caused by MiD49/MiD51 KO.
Fig. 8: Oleic acid-induced mitochondrial fission is MiD-dependent.

Similar content being viewed by others

Data availability

Raw numerical data for all quantifications have been provided in Statistical Source Data. Unprocessed blots and gels for all relevant figures have been provided in Image Source Data. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Chan, D. C. Mitochondrial dynamics and its involvement in disease. Annu. Rev. Pathol. Mech. Dis. 15, 235–259 (2020).

    Article  CAS  Google Scholar 

  2. Kraus, F., Roy, K., Pucadyil, T. J. & Ryan, M. T. Function and regulation of the divisome for mitochondrial fission. Nature 590, 57–66 (2021). vol.

    Article  CAS  PubMed  Google Scholar 

  3. Ngo, J. et al. Mitochondrial morphology controls fatty acid utilization by changing CPT1 sensitivity to malonyl‐CoA. EMBO J. https://doi.org/10.15252/embj.2022111901 (2023).

  4. Pickles, S., Vigié, P. & Youle, R. J. Mitophagy and quality control mechanisms in mitochondrial maintenance. Curr. Biol. 28, R170–R185 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. König, T. et al. MIROs and DRP1 drive mitochondrial-derived vesicle biogenesis and promote quality control. Nat. Cell Biol. 23, 1271–1286 (2021).

    Article  PubMed  Google Scholar 

  6. Fröhlich, C. et al. Structural insights into oligomerization and mitochondrial remodelling of dynamin 1-like protein. EMBO J. 32, 1280–1292 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Koirala, S. et al. Interchangeable adaptors regulate mitochondrial dynamin assembly for membrane scission. Proc. Natl Acad. Sci. USA 110, 1342–1351 (2013).

    Article  Google Scholar 

  8. Bui, H. T. & Shaw, J. M. Dynamin assembly strategies and adaptor proteins in mitochondrial fission. Curr. Biol. 23, 891–899 (2013).

    Article  Google Scholar 

  9. Gandre-Babbe, S. & van der Bliek, A. M. The novel tail-anchored membrane protein Mff controls mitochondrial and peroxisomal fission in mammalian cells. Mol. Biol. Cell 19, 2402–2412 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Otera, H. et al. Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. J. Cell Biol. 191, 1141–1158 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Losón, O. C., Song, Z., Chen, H. & Chan, D. C. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol. Biol. Cell 24, 659–667 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Shen, Q. et al. Mutations in Fis1 disrupt orderly disposal of defective mitochondria. Mol. Biol. Cell 25, 145–159 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Otera, H., Miyata, N., Kuge, O. & Mihara, K. Drp1-dependent mitochondrial fission via MiD49/51 is essential for apoptotic cristae remodeling. J. Cell Biol. 212, 531–544 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Osellame, L. D. et al. Cooperative and independent roles of the Drp1 adaptors Mff, MiD49 and MiD51 in mitochondrial fission. J. Cell Sci. 129, 2170–2181 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Palmer, C. S. et al. MiD49 and MiD51, new components of the mitochondrial fission machinery. EMBO Rep. 12, 565–573 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhao, J. et al. Human MIEF1 recruits Drp1 to mitochondrial outer membranes and promotes mitochondrial fusion rather than fission. EMBO J. 30, 2762–2778 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Palmer, C. S. et al. Adaptor proteins MiD49 and MiD51 can act independently of Mff and Fis1 in Drp1 recruitment and are specific for mitochondrial fission. J. Biol. Chem. 288, 27584–27593 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Elgass, K. D., Smith, E. A., LeGros, M. A., Larabell, C. A. & Ryan, M. T. Analysis of ER-mitochondria contacts using correlative fluorescence microscopy and soft X-ray tomography of mammalian cells. J. Cell Sci. https://doi.org/10.1242/jcs.169136 (2015).

  19. Richter, V. et al. Structural and functional analysis of mid51, a dynamin receptor required for mitochondrial fission. J. Cell Biol. https://doi.org/10.1083/jcb.201311014 (2014).

  20. Losón, O. C. et al. The mitochondrial fission receptor MiD51 requires ADP as a cofactor. Structure 22, 367–377 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Losõn, O. C. et al. Crystal structure and functional analysis of MiD49, a receptor for the mitochondrial fission protein Drp1. Protein Sci. https://doi.org/10.1002/pro.2629 (2015).

  22. Clinton, R. W., Francy, C. A., Ramachandran, R., Qi, X. & Mears, J. A. Dynamin-related protein 1 oligomerization in solution impairs functional interactions with membrane-anchored mitochondrial fission factor. J. Biol. Chem. 291, 478–492 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Kamerkar, S. C., Kraus, F., Sharpe, A. J., Pucadyil, T. J. & Ryan, M. T. Dynamin-related protein 1 has membrane constricting and severing abilities sufficient for mitochondrial and peroxisomal fission. Nat. Commun. 9, 1–15 (2018).

    Article  Google Scholar 

  24. Liu, A., Kage, F. & Higgs, H. N. Mff oligomerization is required for Drp1 activation and synergy with actin filaments during mitochondrial division. Mol. Biol. Cell 32, ar5 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ji, W. K., Hatch, A. L., Merrill, R. A., Strack, S. & Higgs, H. N. Actin filaments target the oligomeric maturation of the dynamin GTPase Drp1 to mitochondrial fission sites. eLife 4, e11553 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Hatch, A. L., Ji, W. K., Merrill, R. A., Strack, S. & Higgs, H. N. Actin flaments as dynamic reservoirs for Drp1 recruitment. Mol. Biol. Cell 27, 3109–3121 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Korobova, F., Ramabhadran, V. & Higgs, H. N. An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science 339, 464–467 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Chakrabarti, R. et al. INF2-mediated actin polymerization at the ER stimulates mitochondrial calcium uptake, inner membrane constriction, and division. J. Cell Biol. 217, 251–268 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liesa, M. & Shirihai, O. S. Mitochondrial dynamics in the regulation of nutrient utilization and energy expenditure. Cell Metab. 17, 491–506 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Houten, S. M., Violante, S., Ventura, F. V. & Wanders, R. J. A. The biochemistry and physiology of mitochondrial fatty acid β-oxidation and its genetic disorders. Annu. Rev. Physiol. 78, 23–44 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Gonzalez-Baro, M. R. & Coleman, R. A. Mitochondrial acyltransferases and glycerophospholipid metabolism. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1862, 49–55 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Irifune, H. et al. GPAM mediated lysophosphatidic acid synthesis regulates mitochondrial dynamics in acute myeloid leukemia. Cancer Sci. 114, 3247–3258 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Constantinides, P. P. & Steim, J. M. Physical properties of fatty acyl-CoA. Critical micelle concentrations and micellar size and shape. J. Biol. Chem. 260, 7573–7580 (1985).

    Article  CAS  PubMed  Google Scholar 

  34. Tranchant, T. et al. Long-term supplementation of culture medium with essential fatty acids alters α-linolenic acid uptake in Caco-2 clone TC7. Can. J. Physiol. Pharmacol. 76, 621–629 (1998).

    CAS  PubMed  Google Scholar 

  35. Yang, X., Ma, Y., Li, N., Cai, H. & Bartlett, M. G. Development of a method for the determination of acyl-CoA compounds by liquid chromatography mass spectrometry to probe the metabolism of fatty acids. Anal. Chem. 89, 813–821 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Davda, D. et al. Profiling targets of the irreversible palmitoylation inhibitor 2-bromopalmitate. ACS Chem. Biol. 8, 1912–1917 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pinkosky, S. L. et al. Long-chain fatty acyl-CoA esters regulate metabolism via allosteric control of AMPK β1 isoforms. Nat. Metab. 2, 873–881 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Noel, R. J., Antinozzi, P. A., McGarry, J. D. & Newgard, C. B. Engineering of glycerol-stimulated insulin secretion in islet β cells. Differential metabolic fates of glucose and glycerol provide insight into mechanisms of stimulus-secretion coupling. J. Biol. Chem. 272, 18621–18627 (1997).

    Article  CAS  PubMed  Google Scholar 

  39. Kalia, R. et al. Structural basis of mitochondrial receptor binding and constriction by DRP1. Nature 558, 401–405 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Manor, U. et al. A mitochondria-anchored isoform of the actin-nucleating spire protein regulates mitochondrial division. eLife 4, e08828 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Liu, R. & Chan, D. C. The mitochondrial fission receptor Mff selectively recruits oligomerized Drp1. Mol. Biol. Cell 26, 4466–4477 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kleele, T. et al. Distinct fission signatures predict mitochondrial degradation or biogenesis. Nature 593, 435–439 (2021).

    Article  CAS  PubMed  Google Scholar 

  43. Zhu, Y. et al. Carnitine palmitoyltransferase 1A promotes mitochondrial fission by enhancing Mff succinylation in ovarian cancer. Commun. Biol. 6, 1–14 (2023).

    Article  Google Scholar 

  44. He, J. et al. The acyl-CoA-binding protein Acb1 regulates mitochondria, lipid droplets, and cell proliferation. FEBS Lett. 596, 1795–1808 (2022).

    Article  CAS  PubMed  Google Scholar 

  45. Hunkeler, M. et al. Structural basis for regulation of human acetyl-CoA carboxylase. Nature 558, 470–474 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Spudich, J. A. & Watt, S. The regulation of rabbit skeletal muscle contraction. I. Biochemical studies of the interaction of the tropomyosin-troponin complex with actin and the proteolytic fragments of myosin. J. Biol. Chem. 246, 4866–4871 (1971).

    Article  CAS  PubMed  Google Scholar 

  47. Woldegiorgis, G., Spennetta, T., Corkey, B. E., Williamson, J. R. & Shrago, E. Extraction of tissue long-chain acyl-CoA esters and measurement by reverse-phase high-performance liquid chromatography. Anal. Biochem. 150, 8–12 (1985).

    Article  CAS  PubMed  Google Scholar 

  48. Booth, D. S., Avila-Sakar, A. & Cheng, Y. Visualizing proteins and macromolecular complexes by negative stain EM: from grid preparation to image acquisition. J. Vis. Exp. 58, e3227 (2011).

    Google Scholar 

  49. Kikumoto, M. & Oosawa, F. Thermodynamic measurements of actin polymerization with various cation species. Cytoskeleton 74, 465–471 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Delgado, S. Liu, Z. Svindrych and A. I. Ferreira Verissimo for much appreciated help during these investigations. We greatly value the efforts of M. Ryan and A. Sharpe for providing MiD KO cells made on the other side of the world. We also thank A. Divakaruni and A. Jones for advice on acyl-CoA extraction methods from cells, and Z. Comeny for working so well in transferring materials. This work was supported by the National Institutes of Health (NIH) R35 GM122545 and R01 DK088826 to H.N.H., NIH R35 GM150942 and a Boettcher Foundation Webb-Waring Biomedical Research Award to H.A., NIH P20 GM113132 to the Dartmouth BioMT and by the Deutsche Forschungsgemeinschaft fellowship KA5106/1-1 (418076373) to F.K.

Author information

Authors and Affiliations

Authors

Contributions

A.L. was the main experimentalist for the biochemical experiments (Figs. 14 and associated Extended Data figures). F.K. was the main experimentalist for the cellular experiments (Figs. 58 and associated Extended Data figures.). H.N.H. conducted acyl-CoA:MiD ratio experiments, RT–PCR analysis and CMC determination. G.S. and H.A. conducted negative-stain EM experiments. A.F.A. assisted with acyl-CoA binding preference experiments. M.P.H.A. assisted with mutant MiD51 biochemical analysis. A.L., F.K. and H.N.H. discussed the experimental approach and results and wrote/edited the paper (including figure preparation).

Corresponding author

Correspondence to Henry N. Higgs.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Christoph Thiele and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Proteins used in this study, and effect of palmitoyl-CoA on MiD49 oligomerization.

a, Bar diagrams of MiD49 and MiD51, showing the regions used in our biochemical studies: 125–454 for murine MiD49 (38.7 kDa), and 134-463 for human MiD51 (38.3 kDa). TM = transmembrane sequence, DR = disordered region. Green region = segment used for biochemical studies here, and for crystallization studies by others. b, Coomassie-stained SDS–PAGE of proteins used in this study. 2 µg protein loaded. Mass markers in kDa shown. Protein preparations were conducted independently with similar outcomes 3 times for MiD49 and MiD51-WT, and 2 times for MiD51-R342A and MiD51-Y185A. c, Superose 6 size-exclusion chromatography of MiD49 cytoplasmic region (100 µM) mixed with varying concentrations of palmitoyl-CoA. d, Velocity analytical ultracentrifugation of peak fractions from Superose 6 size-exclusion chromatography of MiD49 cytoplasmic region (100 µM) incubated without or with palmitoyl-CoA (100 µM). Molecular masses calculated by these vAUC data: 36.4 kDa (without palmitoyl-CoA), and 4815 kDa (with palmitoyl-CoA). Unprocessed gels are available in source data.

Extended Data Fig. 2 Negative-stain EM of MiD49 and MiD51 oligomers.

Peak oligomer fractions from Superose 6 chromatography after mixing MiD49 (100 µM) or MiD51 (50 µM) with palmitoyl-CoA (50 or 500 µM, respectively). a, Full-field examples for MiD49 and MiD51. Scale bars, 100 nm. b, Graph of long axis and short axis lengths for 18 particles of MiD49 or MiD51 oligomers. c, Montage of MiD49 (left) or MiD51 (right) oligomer particles. Scale bars, 50 nm. d, Palmitoyl-CoA alone (1 mM) in same buffer as MiDs (10 mM Hepes pH 7.4, 65 mM KCl, 1 mM MgCl2, 1 mM EGTA, 1 mM DTT). Scale bars: 100 nm (main panel), 50 nm (inset). e, Graph of particles/field (800 ×800 nm field) versus palmitoyl-CoA concentration. 3–5 fields quantified for each concentration. X intercept for linear fit of the 125, 250, 500, and 1000 μM data points (after accounting for background particles, 8.2/field) represents critical micelle concentration (cmc, 47 µM). Inset shows data points closer to the cmc (red line indicates mean of 0 μM palmitoyl-CoA particles).

Extended Data Fig. 3 Palmitoyl-CoA quantification in MiD49 oligomer fraction.

a, Reversed-phase HPLC profile of 1.5 nmole palmitoyl-CoA (red) or solvent alone (blue). Dashed grey line represents acetonitrile concentration. b, Palmitoyl-CoA peaks from four quantities of loaded palmitoyl-CoA (0, 0.5, 1, 1.5 and 2 nmole). c, Peak area as a function of palmitoyl-CoA loaded. d, Palmitoyl-CoA HPLC peak obtained from 200 mL of MiD49 oligomer fraction (Superose 6 oligomer fraction after incubating 100 µM MiD49 with 500 µM palmitoyl-CoA). Peak area, 3.42 A.U.-min, corresponding to 1.1 nmole (5.5 µM) palmitoyl-CoA. MiD49 concentration in this fraction (by Bradford assay), 5.3 µM. e, Standard curve of phosphate using the Fiske-Subbarow assay. f, Palmitoyl-CoA concentrations calculated from Fiske-Subbarow assay in which 400 µL oligomer fraction from Superose 6 was assayed. ‘Condition’ refers to the µM amount of palmitoyl-CoA that was incubated with 100 µM MiD49 (37˚C, 1 hr) prior to separation of oligomers from monomers by Superose 6. MiD49 concentration determined by Bradford assay.

Extended Data Fig. 4 HPLC analysis of acyl-CoAs bound to MiD49 oligomer.

a, Individual HPLC traces of each of the acyl-CoAs (4 nmole loaded). b, HPLC trace of the MiD49 oligomer fraction from the following experiment. MiD49 cytoplasmic construct (100 µM) was mixed simultaneously with 83.3 µM of six acyl-CoAs (stearoyl, oleoyl, palmitoyl, myristoyl, lauroyl, octanoyl) for 1 hr at 37˚C, then MiD49 oligomer was isolated by Superose 6 chromatography. Acyl-CoA distribution was then analysed in the oligomer fraction by reversed-phase HPLC. The Y axis label of A.U. in all graphs in this figure represents mA.U.

Extended Data Fig. 5 Effects of LCACA on MiD51 biochemical properties.

a-c, Effect of MiD49 and actin filaments on DRP1 activity. a, DRP1 GTPase assays (0.75 µM DRP1) alone (black points) or in the presence of 0.5 µM actin filaments (red). b, DRP1 GTPase assays (0.75 µM DRP1) alone (black points) or in the presence of 10 µM Mff alone (grey) or with 250 nM MiD49 oligomers (red) or monomers (blue) added. c, Effect of varying concentrations of Mff on DRP1 GTPase activity (0.75 µM DRP1) in the absence or presence of 250 nM MiD49 oligomers (red) or monomers (blue). Full curve to 100 µM Mff. Zoom to 20 µM Mff shown in Fig. 2f. d, Blue-native gel electrophoresis of MiD51 cytoplasmic region (50 µM) mixed with 500 µM of the indicated molecules. e, Size-exclusion chromatography (Superose 6) of MiD51 (50 µM) alone (blue) or in the presence of 100 µM palmitoyl-CoA (red). f, Velocity analytical ultracentrifugation of the MiD51 oligomer peak Fig. 3b (left) and the monomer MiD51 peak (right), showing calculated molecular masses. g, Blue-native gel electrophoresis of MiD51 (50 µM) with 500 µM of the indicated molecules, or with 500 µM palmitoyl-CoA + the indicated concentrations of ADP, CoA or acetyl-CoA. h, MANT-ADP binding assay in which 300 nM MANT-ADP is mixed with the indicated concentrations of MiD51 or MiD49 and the fluorescence intensity monitored. i, DRP1 GTPase assays containing DRP1 alone (0.75 µM, black points) or in the presence of 0.5 µM MiD51 oligomers (red) or monomers (blue) added. j, Effect of varying concentrations of Mff on DRP1 GTPase activity (0.75 µM DRP1) in the absence or presence of 250 nM MiD51 oligomers (red) or monomers (blue). Full curve to 100 µM Mff. Zoom to 20 µM Mff shown in Fig. 3j. k, Effect of varying concentrations of actin filaments on DRP1 GTPase activity (0.75 µM DRP1) in the absence (black) or presence of MiD51 oligomers (blue). Source numerical data and unprocessed gels are available in source data.

Extended Data Fig. 6 Expression of MiD49 and MiD51-GFP fusion constructs in HeLa cells.

a, C-terminal GFP fusion constructs of MiD49 or MiD51 were transiently transfected into HeLa cells, then cells were fixed and stained for mitochondria (Tom20). At low expression levels (requiring >500 msec exposure at 100% laser power), the GFP fusions did not alter mitochondrial length, and displayed a punctate appearance on mitochondria. At high expression (<100 msec exposure, 50% laser power), the GFP pattern on mitochondria was uniform and mitochondria were hyperfused. Scale bars: 20 µm (whole cell) and 3 µm (insets). Experiment was repeated independently with similar outcomes 3 times. b, Examples of image processing for quantification of GFP-MiD51 puncta size, and % mitochondrial area covered by GFP-MiD51. Boxed regions are shown in higher magnification below overview images. Binary masks of the respective inset images are shown on the right. These binary masks are representative of the images that were used for analyses in Fig. 5 D and E. Scale bars are 10 µm in full images and 3 µm in insets. c, C-terminal GFP fusion constructs of the indicated MiD51 constructs were transiently transfected into HeLa cells, then cells were fixed and stained for mitochondria (Tom20). Cells were selected for high expression under the criteria described above. Images at right represent indicated boxed regions. Examples for WT and Y185 constructs represent hyperfused phenotype quantified in panel d (blue bar), while example for R342A mutant represents collapsed phenotype (grey bar). Scale bars, 20 µm in full images and 3 µm in insets. d, Graph quantifying % cells displaying hyperfused (blue) or collapsed (grey) mitochondria. Representative examples of each pattern are shown in c. N = 122, 82, and 85 cells were analysed for MiD51-WT, Y185A, R342A, respectively. Error bars in graph represent standard error of means. P values were determined by one-sided ANOVA (Dunnett’s multiple comparisons) test. e, RT-PCR analysis of MiD49/51 knockdown in HeLa cells. Left: agarose gels (SybrSafe-stained) of PCR reactions for GAPDH, MiD49 and MiD51 using cDNAs from three independent control KD and three MiD49/MiD51 KD samples. Lengths of PCR products (in base pairs) indicated at left. Right: Bar graph illustrating relative mRNA levels of MiD49 and MiD51 compared to control siRNA-treated samples. Data were quantified from gels and normalized to the GAPDH signals independently for each sample. Error bars represent standard deviations. P values were determined by unpaired two-sided Student’s t-test. Source numerical data are available in source data.

Extended Data Fig. 7 2-bromopalmitate induced mitochondrial fission is MiD-dependent.

a, Fixed-cell fluorescence micrographs of HeLa cells transfected with the indicated siRNAs, treated for 1-hr with either 2-BP or MP and stained for actin (TRITC-phalloidin, red) with Dapi (magenta) and mitochondria (Tom20, green). Experiment was repeated independently with similar outcomes 3 times. b, Quantification of relative mitochondrial lengths between the MP and 2-BP treatments. Data normalized to the MP treatment. Corresponding non-normalized data are in panel c. Error bars in graph represent standard deviations from three independent experiments. P values were determined by unpaired two-sided Student’s t-test. N represents the number of cells analysed per condition (three independent experiments conducted). c, Quantification of mean mitochondrial area between the MP and 2-BP treatments. Corresponding normalized data are in panel b. Box and whisker plots with boxes including 50% (25–75%) and whiskers 80% (10–90%) of all measurements; line within box represents the median and outliers are shown as dots. P values were determined by unpaired two-sided Student’s t-test. N represents the number of cells analysed per condition (three independent experiments conducted). d, Western blot of HeLa cells knocked down for DRP1, confirming the efficiency of the used siRNA. Actin and GAPDH serve as loading controls. Source numerical data and unprocessed blots are available in source data. Experiment was repeated independently with similar outcomes 3 times.

Extended Data Fig. 8 Effects of Triacsin C and Etomoxir on mitochondria morphology.

(a) MEF wild-type cells treated with 5 µM Triacsin C or DMSO for 1 hr were fixed and stained for Tom20 as well as F-actin and Dapi. Boxed regions depict higher magnification images on the right. Scale bars, 20 µm in overview images and 5 µm in zooms. Box and whisker plot on the right showing mean mitochondrial area quantifications for both conditions. Box and whisker plots with boxes including 50% (25–75%) and whiskers 80% (10–90%) of all measurements; line within box represents the median and outliers are shown as dots. P values were determined by unpaired two-sided Student’s t-test. N represents the number of cells analysed per condition (three independent experiments).(b) HeLa cells treated with either 3 or 100 µM Etomoxir (or DMSO as a control) for 1 hr were fixed and stained for Tom20 as well as F-actin and Dapi. Boxed regions depict higher magnification images on the right. Scale bars, 20 µm in overview images and 5 µm in zooms. Box and whisker plot on the right showing mean mitochondrial area quantifications for respective conditions. Box and whisker plots with boxes including 50% (25–75%) and whiskers 80% (10–90%) of all measurements; line within box represents the median and outliers are shown as dots. P values were determined by one-sided ANOVA (Dunnett’s multiple comparisons) test. N represents the number of cells analysed per condition (two independent replicates). Source numerical data are available in source data.

Extended Data Fig. 9 Model for MiD function in activation of fatty acid oxidation.

Step 1. Long-chain fatty acid enters cytoplasm from the extracellular milieu (1 A) or from intracellular lipid droplets (1B). Step 2. Fatty acid is coupled to CoA through fatty acyl-CoA synthetase. Step 3. Fatty acyl-CoA binds to MiD49 and/or MiD51 monomers on the outer mitochondrial membrane (OMM), inducing their oligomerization. Step 4. Oligomerized MiD initiates assembly of active DRP1 oligomers on the OMM. Step 5. DRP1 oligomerization continues on the OMM to create a ring around the mitochondrion. This step might be facilitated (or require) Mff. Step 6. DRP1-mediated constriction of the OMM. Step 7. mitochondrial fission.

Supplementary information

Supplementary Information

Supplementary Video legends 1–4.

Reporting Summary

Peer Review File

Supplementary Video 1

MiD49–GFP dynamics. HeLa cell co-transfected with MiD49–GFP (green) and Mito-Plum (red). Scale bar in whole cell is 10 µm and 3 µm in inset.

Supplementary Video 2

MiD51-WT GFP dynamics. HeLa cell co-transfected with MiD51–GFP (green) and Mito-Plum (red). Scale bar in whole cell is 10 µm and 3 µm in inset.

Supplementary Video 3

MiD51-Y185A GFP dynamics. HeLa cell co-transfected with MiD51-Y185A–GFP (green) and Mito-Plum (red). Scale bar in whole cell is 10 µm and 3 µm in inset.

Supplementary Video 4

MiD51-R342A GFP dynamics. HeLa cell co-transfected with MiD51-R342A–GFP (green) and Mito-Plum (red). Scale bar in whole cell is 10 µm and 3 µm in inset.

Source data

Statistical Source Data

Statistical source data.

Image Source Data

Unprocessed western blots and gels.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, A., Kage, F., Abdulkareem, A.F. et al. Fatty acyl-coenzyme A activates mitochondrial division through oligomerization of MiD49 and MiD51. Nat Cell Biol (2024). https://doi.org/10.1038/s41556-024-01400-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41556-024-01400-3

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