Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2017 Mar 13;114(12):E2268–E2269. doi: 10.1073/pnas.1618610114

Reply to Filadi et al.: Does Mitofusin 2 tether or separate endoplasmic reticulum and mitochondria?

Deborah Naon a,b, Marta Zaninello a,c,d, Marta Giacomello a,c, Tatiana Varanita a,c, Francesca Grespi a,c, Sowmya Lakshminaranayan a,d, Annalisa Serafini a,c, Martina Semenzato a,c, Stephanie Herkenne a,c, Maria Isabel Hernández-Alvarez e, Antonio Zorzano e, Diego De Stefani b, Gerald W Dorn II f, Luca Scorrano a,c,1
PMCID: PMC5373396  PMID: 28289205

We thank Filadi et al. for their comments (1) on our paper (2), where we address whether the discrepancies between their paper (3) and our original discovery of Mitofusin (Mfn) 2 as an endoplasmic reticulum (ER)–mitochondria tether (4) resulted from: (i) clonal effects of chronic Mfn2 ablation, (ii) proximity measurement inappropriateness, or (iii) changes in mitochondrial Ca2+ uniporter (MCU) levels in WT and Mfn2−/−cells. Filadi et al. (1) conclude that we fell short in solving the issue and that our data reinforce Mfn2 function as an ER–mitochondria spacer (3).

First, Filadi et al. (1) reason that we did not measure contacts number upon Mfn2 ablation. However, contact surface (which depends on contact number and extent) can be extracted from the ER–mitochondria contact coefficient and data in our paper (2). The average mitochondrial surface contacting ER is: WT cells, 7.9%; Mfn1−/−, 8.4%; Mfn2−/−, 5.9%; Mfn1,2−/−, 5.0% (data from table S1 in ref. 2). Also using data from tables S2 and S3 in our paper (2), we conclude that Mfn2 ablation decreases the ER-contacting mitochondrial surface by ∼20–35%, potentially explaining the confocal microscopy juxtaposition reduction.

Second, Filadi et al. (1) question conclusions based on fluorescent organelle proximity probes. ddGFP and FRET-based indicator of ER–mitochondria proximity (FEMP) do not artificially juxtapose organelles: ddGFP KOFF is >> KON, implying that dimerization depends on proximity and not vice versa (5); FEMP does not spontaneously and stably dimerize, as confirmed by its response to rapamycin (see ref. 6 and figure S2 in ref. 2). Mathematically, the lower FRET ratio upon Mfn2 ablation (figures 1 and 2 in ref. 2) results from lower FRETbasal and FRETmaximal values (Tables 1 and 2), not from increased FRETmaximal. Thus, ddGFP and FEMP are reliable organelle proximity sensors.

Table 1.

Basal and maximal FEMP FRET values upon Mfn2 ablation: Figure 1H in ref. 2

WT Mfn2−/−
FRETbasal FRETmaximal FRETbasal FRETmaximal
1.1 ± 0.05 1.7 ± 0.01 0.76 ± 0.01* 1.03 ± 0.02*
*

P < 0.005 in a two-tailed Student’s t test vs. WT FRETbasal or FRETmaximal.

Table 2.

Basal and maximal FEMP FRET values upon Mfn2 ablation: Figure 2A in ref. 2

Scr Mfn2shRNA1 Mfn2shRNA2
FRETbasal FRETmaximal FRETbasal FRETmaximal FRETbasal FRETmaximal
0.78 ± 0.05 1.43 ± 0.04 0.66 ± 0.02* 0.83 ± 0.01* 0.67 ± 0.01 0.85 ± 0.01*
*

P < 0.005 in a two-tailed Student’s T test vs. Scr FRETbasal or FRETmaximal.

Finally, Filadi et al. (1) raise technical concerns on presented data. First, in the same experimental conditions, mitochondrial Ca2+ peak does not span two orders-of-magnitude as stated in their letter (1): it is 160 nM in figure 3B of ref. 2 and 390 ± 150 nM in figure 3C of ref. 2 (average of five independent experiments ± SEM). Panel F of figure 3 in ref. 2 cannot be compared with panels A and B because conditions were different (as described in the legend to the figure): Cre-infected Mfn2flx/flx cells were preincubated in Ca2+-free media to equalize cytosolic Ca2+ peaks (figure 3 D and E of ref. 2). Second, we excluded respiration defects in purified Mfn2 liver knockout mitochondria (Mfn2LKO; figure S4 of ref. 2) that, as suggested by Filadi et al. (1), could limit mitochondrial Ca2+ uptake in Mg2+-free media. Third, mitochondrial Ca2+ uptake rates are not “clearly slower” in Mfn2LKO mitochondria (1), but superimposable to the WT ones (figure 3 I–K in ref. 2; WT: 11.3 ± 0.6, Mfn2LKO: 11.3 ± 0.9 s−1). Fourth, in WT cells, MCU levels are indeed affected by density (1), but at confluency are lower than in Mfn2−/− cells (figure S5 of ref. 2) and not vice versa (3). Mitochondrial Ca2+ transients are lesser in Mfn2−/− cells even upon MCU overexpression (figure 5 D and E of ref. 2): reduced MCU levels cannot therefore explain the decreased mitochondrial Ca2+ uptake in Mfn2−/− cells.

The careful Filadi et al. analysis (1, 3) highlights the ER–mitochondria interface complexity. We maintain that our acute Mfn2 genetic deletion experiments, reliable organelle proximity probes, and Ca2+ measurements (2) address the raised issues in their letter (3) and add to multiple independent papers reporting ER–mitochondria tethering by Mfn2 (4, 5, 710). A deeper knowledge of the ER–mitochondria interface architecture could help resolve this controversy.

Footnotes

The authors declare no conflict of interest.

References

  • 1.Filadi R, et al. On the role of Mitofusin 2 in endoplasmic reticulum–mitochondria tethering. Proc Natl Acad Sci USA. 2017;114:E2266–E2267. doi: 10.1073/pnas.1616040114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Naon D, et al. Critical reappraisal confirms that Mitofusin 2 is an endoplasmic reticulum–mitochondria tether. Proc Natl Acad Sci USA. 2016;113(40):11249–11254. doi: 10.1073/pnas.1606786113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Filadi R, et al. Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Natl Acad Sci USA. 2015;112(17):E2174–E2181. doi: 10.1073/pnas.1504880112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.de Brito OM, Scorrano L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature. 2008;456(7222):605–610. doi: 10.1038/nature07534. [DOI] [PubMed] [Google Scholar]
  • 5.Alford SC, Ding Y, Simmen T, Campbell RE. Dimerization-dependent green and yellow fluorescent proteins. ACS Synth Biol. 2012;1(12):569–575. doi: 10.1021/sb300050j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Csordás G, et al. Structural and functional features and significance of the physical linkage between ER and mitochondria. J Cell Biol. 2006;174(7):915–921. doi: 10.1083/jcb.200604016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen Y, et al. Mitofusin 2-containing mitochondrial-reticular microdomains direct rapid cardiomyocyte bioenergetic responses via interorganelle Ca(2+) crosstalk. Circ Res. 2012;111(7):863–875. doi: 10.1161/CIRCRESAHA.112.266585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schneeberger M, et al. Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell. 2013;155(1):172–187. doi: 10.1016/j.cell.2013.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Sugiura A, et al. MITOL regulates endoplasmic reticulum-mitochondria contacts via Mitofusin2. Mol Cell. 2013;51(1):20–34. doi: 10.1016/j.molcel.2013.04.023. [DOI] [PubMed] [Google Scholar]
  • 10.Li D, Li X, Guan Y, Guo X. Mitofusin-2-mediated tethering of mitochondria and endoplasmic reticulum promotes cell cycle arrest of vascular smooth muscle cells in G0/G1 phase. Acta Biochim Biophys Sin (Shanghai) 2015;47(6):441–450. doi: 10.1093/abbs/gmv035. [DOI] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES