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.

  • Letter
  • Published:

Molecular basis of selective mitochondrial fusion by heterotypic action between OPA1 and cardiolipin

Abstract

Mitochondria are highly dynamic organelles that undergo frequent fusion and fission. Optic atrophy 1 (OPA1) is an essential GTPase protein for both mitochondrial inner membrane (IM) fusion and cristae morphology1,2. Under mitochondria-stress conditions, membrane-anchored L-OPA1 is proteolytically cleaved to form peripheral S-OPA1, leading to the selection of damaged mitochondria for mitophagy2,3,4. However, molecular details of the selective mitochondrial fusion are less well understood. Here, we showed that L-OPA1 and cardiolipin (CL) cooperate in heterotypic mitochondrial IM fusion. We reconstituted an in vitro membrane fusion reaction using purified human L-OPA1 protein expressed in silkworm, and found that L-OPA1 on one side of the membrane and CL on the other side are sufficient for fusion. GTP-independent membrane tethering through L-OPA1 and CL primes the subsequent GTP-hydrolysis-dependent fusion, which can be modulated by the presence of S-OPA1. These results unveil the most minimal intracellular membrane fusion machinery. In contrast, independent of CL, a homotypic trans-OPA1 interaction mediates membrane tethering, thereby supporting the cristae structure. Thus, multiple OPA1 functions are modulated by local CL conditions for regulation of mitochondrial morphology and quality control.

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

Figure 1: Preparation of recombinant human OPA1 proteins using silkworm larvae.
Figure 2: L-OPA1-dependent membrane fusion.
Figure 3: Heterotypic membrane fusion via opposed L-OPA1 and CL.
Figure 4: Specific L-OPA1 to CL interaction is required for membrane fusion.
Figure 5: Role of S-OPA1 in L-OPA1-mediated membrane fusion.

Similar content being viewed by others

References

  1. Friedman, J. R. & Nunnari, J. Mitochondrial form and function. Nature 505, 335–343 (2014).

    Article  CAS  Google Scholar 

  2. Pernas, L. & Scorrano, L. Mito-morphosis: mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu. Rev. Physiol. 78, 505–531 (2016).

    Article  CAS  Google Scholar 

  3. MacVicar, T. & Langer, T. OPA1 processing in cell death and disease—the long and short of it. J. Cell. Sci. 129, 2297–2306 (2016).

    Article  CAS  Google Scholar 

  4. Ishihara, N., Fujita, Y., Oka, T. & Mihara, K. Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J. 25, 2966–2977 (2006).

    Article  CAS  Google Scholar 

  5. McNew, J. A., Sondermann, H., Lee, T., Stern, M. & Brandizzi, F. GTP-dependent membrane fusion. Annu. Rev. Cell. Dev. Biol. 29, 529–550 (2013).

    Article  CAS  Google Scholar 

  6. Delettre, C. et al. Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy. Nat. Genet. 26, 207–210 (2000).

    Article  CAS  Google Scholar 

  7. Anand, R. et al. The i-AAA protease YME1L and OMA1 cleave OPA1 to balance mitochondrial fusion and fission. J. Cell. Biol. 204, 919–929 (2014).

    Article  CAS  Google Scholar 

  8. Griparic, L., van der Wel, N. N., Orozco, I. J., Peters, P. J. & van der Bliek, A. M. Loss of the intermembrane space protein Mgm1/OPA1 induces swelling and localized constrictions along the lengths of mitochondria. J. Biol. Chem. 279, 18792–18798 (2004).

    Article  CAS  Google Scholar 

  9. Frezza, C. et al. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 126, 177–189 (2006).

    Article  CAS  Google Scholar 

  10. McQuibban, G. A., Saurya, S. & Freeman, M. Mitochondrial membrane remodelling regulated by a conserved rhomboid protease. Nature 423, 537–541 (2003).

    Article  CAS  Google Scholar 

  11. Herlan, M., Bornhovd, C., Hell, K., Neupert, W. & Reichert, A. S. Alternative topogenesis of Mgm1 and mitochondrial morphology depend on ATP and a functional import motor. J. Cell. Biol. 165, 167–173 (2004).

    Article  CAS  Google Scholar 

  12. DeVay, R. M. et al. Coassembly of Mgm1 isoforms requires cardiolipin and mediates mitochondrial inner membrane fusion. J. Cell. Biol. 186, 793–803 (2009).

    Article  CAS  Google Scholar 

  13. Meeusen, S. et al. Mitochondrial inner-membrane fusion and crista maintenance requires the dynamin-related GTPase Mgm1. Cell 127, 383–395 (2006).

    Article  CAS  Google Scholar 

  14. Kajikawa, M. et al. Efficient silkworm expression of human GPCR (nociceptin receptor) by a Bombyx mori bacmid DNA system. Biochem. Biophys. Res. Commun. 385, 375–379 (2009).

    Article  CAS  Google Scholar 

  15. Ardail, D. et al. Mitochondrial contact sites. Lipid composition and dynamics. J. Biol. Chem. 265, 18797–18802 (1990).

    CAS  PubMed  Google Scholar 

  16. Meglei, G. & McQuibban, G. A. The dynamin-related protein Mgm1p assembles into oligomers and hydrolyzes GTP to function in mitochondrial membrane fusion. Biochemistry 48, 1774–1784 (2009).

    Article  CAS  Google Scholar 

  17. Oh, K. J. et al. Conformational changes in BAK, a pore-forming proapoptotic Bcl-2 family member, upon membrane insertion and direct evidence for the existence of BH3-BH3 contact interface in BAK homo-oligomers. J. Biol. Chem. 285, 28924–28937 (2010).

    Article  CAS  Google Scholar 

  18. Song, Z., Ghochani, M., McCaffery, J. M., Frey, T. G. & Chan, D. C. Mitofusins and OPA1 mediate sequential steps in mitochondrial membrane fusion. Mol. Biol. Cell. 20, 3525–3532 (2009).

    Article  CAS  Google Scholar 

  19. Claypool, S. M. Cardiolipin, a critical determinant of mitochondrial carrier protein assembly and function. Biochim. Biophys. Acta 1788, 2059–2068 (2009).

    Article  CAS  Google Scholar 

  20. Lutter, M. et al. Cardiolipin provides specificity for targeting of tBid to mitochondria. Nat. Cell. Biol. 2, 754–761 (2000).

    Article  CAS  Google Scholar 

  21. Choi, S. Y. et al. A common lipid links Mfn-mediated mitochondrial fusion and SNARE-regulated exocytosis. Nat. Cell. Biol. 8, 1255–1262 (2006).

    Article  CAS  Google Scholar 

  22. Chu, C. T. et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat. Cell. Biol. 15, 1197–1205 (2013).

    Article  CAS  Google Scholar 

  23. Weber, T. et al. SNAREpins: minimal machinery for membrane fusion. Cell 92, 759–772 (1998).

    Article  CAS  Google Scholar 

  24. Mima, J. & Wickner, W. Complex lipid requirements for SNARE- and SNARE chaperone-dependent membrane fusion. J. Biol. Chem. 284, 27114–27122 (2009).

    Article  CAS  Google Scholar 

  25. Koshiba, T. et al. Structural basis of mitochondrial tethering by mitofusin complexes. Science 305, 858–862 (2004).

    Article  CAS  Google Scholar 

  26. Ishihara, N., Eura, Y. & Mihara, K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J. Cell. Sci. 117, 6535–6546 (2004).

    Article  CAS  Google Scholar 

  27. Orso, G. et al. Homotypic fusion of ER membranes requires the dynamin-like GTPase atlastin. Nature 460, 978–983 (2009).

    Article  CAS  Google Scholar 

  28. Patten, D. A. et al. OPA1-dependent cristae modulation is essential for cellular adaptation to metabolic demand. EMBO J. 33, 2676–2691 (2014).

    Article  CAS  Google Scholar 

  29. Ban, T., Heymann, J. A., Song, Z., Hinshaw, J. E. & Chan, D. C. OPA1 disease alleles causing dominant optic atrophy have defects in cardiolipin-stimulated GTP hydrolysis and membrane tubulation. Hum. Mol. Genet. 19, 2113–2122 (2010).

    Article  CAS  Google Scholar 

  30. Schlame, M. & Ren, M. The role of cardiolipin in the structural organization of mitochondrial membranes. Biochim. Biophys. Acta 1788, 2080–2083 (2009).

    Article  CAS  Google Scholar 

  31. Mishra, P., Carelli, V., Manfredi, G. & Chan, D. C. Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation. Cell. Metab. 19, 630–641 (2014).

    Article  CAS  Google Scholar 

  32. Eckert, D. M. & Kim, P. S. Mechanisms of viral membrane fusion and its inhibition. Annu. Rev. Biochem. 70, 777–810 (2001).

    Article  CAS  Google Scholar 

  33. Ban, T. & Ishihara, N. Expression and purification of recombinant human L-OPA1 using BmNPV bacmid-silkworm expression system. Protoc. Exch. http://dx.doi.org/10.1038/protex.2017.053 (2017).

  34. Mima, J., Hickey, C. M., Xu, H., Jun, Y. & Wickner, W. Reconstituted membrane fusion requires regulatory lipids, SNAREs and synergistic SNARE chaperones. EMBO J. 27, 2031–2042 (2008).

    Article  CAS  Google Scholar 

  35. Sugiura, S. & Mima, J. Physiological lipid composition is vital for homotypic ER membrane fusion mediated by the dynamin-related GTPase Sey1p. Sci. Rep. 6, 20407 (2016).

    Article  CAS  Google Scholar 

  36. Eura, Y., Ishihara, N., Yokota, S. & Mihara, K. Two mitofusin proteins, mammalian homologues of FZO, with distinct functions are both required for mitochondrial fusion. J. Biochem. 134, 333–344 (2003).

    Article  CAS  Google Scholar 

  37. Saita, S. et al. Distinct types of protease systems are involved in homeostasis regulation of mitochondrial morphology via balanced fusion and fission. Genes Cells 21, 408–424 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Mima (Osaka University) for advice on the preparation of assays with proteoliposomes, K. Tabata (Kyushu University) for advice on the silkworm expression system, Kurume University EM laboratory, and H. Yagi (Tottori University) and D. Ozawa (Osaka University) for helping with the EM observations, and W. Nishihira for technical support. This work was supported by JSPS KAKENHI grant numbers 26291044 and 16H01209 (N.I.), and 26840026 (T.B.), MEXT-Supported Program for the Strategic Research Foundation at Private Universities, the Takeda Science Foundation (N.I.), the Ono Medical Research Foundation (N.I.) and the Ishibashi Foundation (T.B.).

Author information

Authors and Affiliations

Authors

Contributions

N.I. and T.B. designed the research. T.B., T.I., H.K., S.S. and A.I. performed the experiments. K.Maenaka established and provided the bacmid expression vector system, and T.O. prepared basic procedures for expression and reconstitution of membrane proteins. K.Mihara supervised the study. N.I. and T.B. wrote the manuscript with the comments of the other authors.

Corresponding author

Correspondence to Naotada Ishihara.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Preparation of recombinant OPA1 and OPA1-dependent fusion in vitro or in vivo.

(a) Procedures for the fractionation and purification of recombinant OPA1. (b,c) SDS-PAGE analysis and Coomassie blue staining of L-OPA1 (b) and S-OPA1 (c). Asterisk: OPA1 protein. Fractionation experiments were carried out more than 10 times for L-OPA1 and more than 3 times for S-OPA1 with similar results. (d) Calculation of fusion activity by fitting to the exponential curve with the trace from the experiments shown in Fig. 2a. In the 1:4,000 condition, 25 L-OPA1 molecules, which might form 2.5 complexes (Fig. 1h) in each liposome (the number of lipid molecules 100,000)35, could mediate membrane fusion. The goodness of fit was evaluated by the R2 value. (e) Preparation of the OPA1 KO cell line using the CRISPR/Cas9 system. Whole cell homogenates from OPA1 WT or KO HeLa cells were prepared and subjected to immunoblot analysis with the indicated antibodies (the experiment was performed one time). (f) OPA1 WT or KO HeLa cells were fixed, permeabilized, subjected to immunofluorescence analysis with antibodies to TOM20 (red), and examined by confocal microscopy (representative images of more than 3 independent experiments with similar results). Magnified images are shown in the boxed regions. Scale bars, 10 μm. (g,h) Heterotypic mitochondrial fusion in HeLa cells. Mitochondrial fusion was analyzed in cell hybrids from cells expressing mitGFP and mitRFP using OPA1-deficient or control HeLa cells. After 6 h culture, fusion efficiency was analyzed by fluorescence microscopy (g) and cell counting (h). n, number of cell hybrids analyzed from 3 independent experiments: WT × WT (n = 30), WT × CRISPR-OPA1 (n = 30), and CRISPR-OPA1 × CRISPR-OPA1 (n = 30). Scale bar: 10 μm. Statistics source data are available in Supplementary Table 1. Unprocessed original scans of blot is shown in Supplementary Fig. 3.

Supplementary Figure 2 CL conditions for regulation of mitochondrial morphology and quality control.

(a) Comparison of the number, length, and unsaturation of acyl chains of CL used in this study. (b) GTP hydrolysis activity of the deletion mutant (Δ581–941) of L-OPA1 (n = 3 independent measurements, data are mean ± s.d.). (cg) KD of CLS1 using siRNA. (c) RNAs were analyzed by RT-PCR (the experiment was performed one time). Nuclear genome-encoded transcripts were analyzed. (d) Fluorescence images of the control and CLS1 KD cells (representative images of more than 3 independent experiments with similar results). Scale bar: 10 μm. (e) Immunoblot analysis of mitochondrial protein levels using the indicated antibodies (the experiment was performed one time). (f,g) Mitochondrial fusion in CLS1 KD cells and OPA1 KD cells. Mitochondrial fusion was analyzed using HeLa cells expressing mitGFP and mitRFP that were co-cultured and fused with the HVJ envelope. After 6 h culture, fusion efficiency was analyzed by fluorescence microscopy. n, number of cell hybrids analyzed from 3 independent experiments: cont × cont (n = 38), CLS1KD × CLS1KD (n = 41), and OPA1KD × OPA1KD (n = 32). Scale bar: 10 μm. Statistics source data are available in Supplementary Table 1. Unprocessed original scans of agarose gel and blots are shown in Supplementary Fig. 3. (h) Model of the isolation of damaged mitochondria from the active mitochondrial network. OPA1 inactivation and CL remodeling are used as markers for the specific selection of damaged mitochondria.

Supplementary Figure 3 Unprocessed original scans of Coomassie-stained gels, agarose gels, and immunoblots shown in the Figures and Supplementary Figures.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1755 kb)

Supplementary Table 1

Supplementary Information (XLSX 57 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ban, T., Ishihara, T., Kohno, H. et al. Molecular basis of selective mitochondrial fusion by heterotypic action between OPA1 and cardiolipin. Nat Cell Biol 19, 856–863 (2017). https://doi.org/10.1038/ncb3560

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3560

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