Sie sind auf Seite 1von 15

Cell Calcium 62 (2017) 1–15

Contents lists available at ScienceDirect

Cell Calcium
journal homepage: www.elsevier.com/locate/ceca

The endoplasmic reticulum-mitochondria coupling in health and


disease: Molecules, functions and significance
Riccardo Filadi a , Pierre Theurey a , Paola Pizzo a,b,∗
a
Department of Biomedical Sciences, University of Padova, Italy
b
Neuroscience Institute, National Research Council (CNR), Padova, Italy

a r t i c l e i n f o a b s t r a c t

Article history: The close apposition between endoplasmic reticulum (ER) and mitochondria represents a key plat-
Received 5 December 2016 form, capable to regulate different fundamental cellular pathways. Among these, Ca2+ signaling and lipid
Received in revised form 9 January 2017 homeostasis have been demonstrated over the last years to be deeply modulated by ER-mitochondria
Accepted 9 January 2017
cross-talk. Given its importance in cell life/death decisions, increasing evidence suggests that alterations
Available online 12 January 2017
of the ER-mitochondria axis could be responsible for the onset and progression of several diseases, includ-
ing neurodegeneration, cancer and obesity. However, the molecular identity of the proteins controlling
Keywords:
this inter-organelle apposition is still debated. In this review, we summarize the main cellular pathways
Calcium (Ca2+ )
Mitochondria
controlled by ER-mitochondria appositions, focusing on the principal molecules reported to be involved
Endoplasmic reticulum (ER) in this interplay and on those diseases for which alterations in organelles communication have been
Inter-organelle contacts reported.
ER-mitochondria tethering © 2017 Elsevier Ltd. All rights reserved.
Mitochondria-Associated Membranes
(MAMs)
Mitofusin 2

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Molecules: proteinaceous structures connecting the ER to mitochondria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Physical and functional tethers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. The elusive role of Mfn2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. Functions: cellular pathways dependent on ER-mitochondria coupling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1. Lipid homeostasis, mitochondria dynamics, autophagy, inflammation and mtDNA distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2. ER-mitochondria Ca2+ cross-talk functionalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Significance: dysfunctions in ER-mitochondria coupling and diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.1. Metabolic syndrome and inflexibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.2. Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
4.3. Alzheimer’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
4.4. Parkinson’s disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
4.5. Amyotrophic lateral sclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
5. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12

∗ Corresponding author at: Department of Biomedical Sciences, University of


Padova, Via U. Bassi 48, 35121 Padova, Italy.
E-mail address: paola.pizzo@unipd.it (P. Pizzo).

http://dx.doi.org/10.1016/j.ceca.2017.01.003
0143-4160/© 2017 Elsevier Ltd. All rights reserved.
2 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

1. Introduction as mitochondrial dynamics, inheritance, protein import, mitochon-


drial DNA (mtDNA) inheritance and mitophagy (see [11] for a recent
Inter-organelle communication represents an emerging aspect review). Importantly, ERMES homologs have not been identified in
in cell biology: indeed, cellular organelles do not function as iso- mammals, while the mitochondrial proteins Miro1/2, involved in
lated structures but rather form dynamic interconnected networks Ca2+ -dependent mitochondrial transport mediated by the kinesin
which can be modulated according to cellular needs. In particular, adapter Milton [12], are the metazoan orthologue of Gem1.
the endoplasmic reticulum (ER)-mitochondria physical/functional The more recently identified tethering complex EMC contains
coupling plays a central role in a variety of cell pathways and in yeast six proteins, Emc1-6 [13] and is also required for phos-
increasing evidence highlights its alteration in several diseases, phatidylserine (PS) import into mitochondria and assembly of
including diabetes and obesity, cancer and neurodegenerative dis- multi-pass ER-membrane proteins [14]. Emc proteins interact with
eases, such as Alzheimer’s Disease (AD), Parkinson’s Disease (PD) the mitochondrial protein Tom5 of the translocase of the outer
and Amyotrophic Lateral Sclerosis (ALS). membrane (TOM) complex, forming a tether between the two
Physical contacts between ER and mitochondria have been organelles [13]. EMC is a highly conserved protein complex, present
firstly observed by EM in the 50’s in rat tissues [1] but such features in every major eukaryotic lineage, and in mammalian cells contains
have been considered for long time to be artifacts of fixation. More four additional proteins (Emc7–10) [15].
recent experiments in living cells expressing GFP variants within Recently, a proteomic analysis identified the ER transmembrane
the two organelles [2] and electron micrograph images of quickly protein Ltc1/Lam6 as a potential additional tether in yeast, thanks
frozen samples [3] have demonstrated conclusively that such phys- to its interaction with the OMM proteins TOM70/71 [16]. Ltc1/Lam6
ical interactions between the two organelles indeed exist (Fig. 1A-B has also been observed to mediate the transfer of sterols between
). Similar structures were then described in yeast and, recently, also the two membranes.
in plants [4,5]. Their composition and thickness, however, are not Less clear is the molecular nature of the physical tether between
constant and the distance between ER and the outer mitochondrial the two organelles in metazoan cells. Several proteins have been
membrane (OMM) can be extremely variable, ranging from ∼10 nm demonstrated to modulate ER-mitochondria coupling: the major-
up to 80–100 nm [6]. Usually, at smooth ER-mitochondria contact ity of them appears to be located on either the OMM or specific ER
sites the gap between the two opposing membranes is smaller membrane domains, called mitochondria-associated membranes
(10–15 nm) than in the case of rough ER (typically 20–30 nm) [7], (MAM) [17]. Despite the growing number of these proteins, the for-
probably to allow ribosomes accommodation. Regions in which mal and univocal demonstration that the lack of a given molecule
OMM and ER membranes proceed in parallel at larger distances abolishes ER-mitochondria contacts has never been provided. Most
(50–100 nm) have been also reported (see for instance [8]), and can likely, different and independent tethering complexes may exist
be continuous to sites in which the two organelles are closer or, on and compensate one for the lack of the others, increasing the com-
the contrary, exist as separate, independent units. However, while plexity of the analysis.
in the case of the closer contacts (below ∼30 nm) [7] electron-dense One multi-protein structure that strongly correlates with func-
filamentous structures, proteinaceous in their nature, clearly pro- tional ER-mitochondria coupling is the IP3R-Grp75-VDAC complex
trude from the two opposing membranes (Fig. 1A), such structures (Fig. 2). In particular, it has been shown that ER resident inosi-
have never been observed for the more distant organelles apposi- tol trisphosphates receptors, IP3Rs, (especially the MAM enriched
tions. Thus, whether these latter are randomly occurring or tightly IP3R3) physically interact with the cytosolic fraction of the mito-
regulated, physiologically relevant events, is still unknown. chondrial chaperone Grp75 and the voltage-dependent anion
This contribution aims to update ER-mitochondria connections channel 1 (VDAC1) of the OMM [18], functionally favoring mito-
from multiple points of view: i) the molecules involved in the for- chondrial Ca2+ uptake upon IP3-dependent ER Ca2+ release (see
mation/modulation of ER-mitochondria inter-organelle structures; below). The role of such complex as a physical tether between the
ii) the specific cellular functions dependent on ER-mitochondria two organelles is however questionable, since DT40 cells knock-out
coupling, especially those relying on their Ca2+ cross-talk; iii) the (KO) for the three IP3R isoforms show, by EM analysis, unmodified
alterations of the ER-mitochondria platform described in different ER-mitochondria physical association [7].
diseases and reported to be pathogenic. The molecular scaffold formed by the ER resident vesicle-
associated membrane protein-associated protein B, VAPB, and
the OMM protein tyrosine phosphatase-interacting protein-51,
2. Molecules: proteinaceous structures connecting the ER PTPIP51, has been also demonstrated to be involved in physical
to mitochondria and functional ER-mitochondria tethering [19,20]. Genetic manip-
ulation of these proteins are linked to variations in organelles
2.1. Physical and functional tethers juxtaposition and altered ER-mitochondria Ca2+ cross-talk. Inter-
estingly, these functionalities are also affected by the expression of
The molecular identity of the structures mediating ER- both VAPB mutants linked to Amyotrophic Lateral Sclerosis (ALS)
mitochondria tethering is more defined in yeast, where two [19] or the ALS-associated proteins TDP43 [20] and FUS [21], that
principal protein complexes are involved: the ER-mitochondria disrupt the endogenous VAPB-PTPIP51 interaction, indicating a key
encounter structure (ERMES) and the ER membrane protein com- role for the mitochondria-ER axis in ALS pathogenesis (see below).
plex (EMC) (Fig. 2). Both structures were identified by genetic In mammalian cells, the mitochondrial protein PTPIP51 has been
screens in S. cerevisiae mutant cells whose growth/lipid metabolism recently reported to physically interact also with the ER-located
defects could be rescued by artificial ER-mitochondria tethers. oxysterol-binding protein (OSBP)-related proteins ORP5 and ORP8
ERMES is formed by the cytosolic protein Mdm12, the ER mem- [22] (see also below), two molecules previously shown to be impor-
brane protein Mmm1 and the OMM proteins Mdm34 and Mdm10 tant for cortical ER-plasma membrane contacts [23].
[9]. In addition to the four core components, the mitochondrial Another example of protein regulating ER-mitochondria jux-
Rho-like GTPase Gem1 represents a facultative regulatory sub- taposition is the phosphofurin acidic cluster sorting 2 protein,
unit of the ERMES complex [10]. ERMES has been functionally PACS-2, a cytosolic multifunctional sorting protein which controls
implicated in ER-mitochondria lipid transport (though different organelles apposition in a B-cell receptor-associated protein 31,
groups failed to find defects in lipid metabolism in cells lacking BAP31 (an ER cargo receptor), −dependent manner [24]. However,
the complex), as well as in several other cellular pathways, such the role of PACS-2 on ER-mitochondria juxtaposition is not com-
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 3

Fig. 1. ER-mitochondria structural apposition. (A) EM image of a FAD-PS2-T122R expressing wt MEF showing some mitochondria (mt) in close apposition to ER membranes.
Scale bar: 500 nm. The inset highlights a close contact in which several proteinaceous structures connecting the two organelles are visible (red arrows). Image acquired by
G. Turacchio, Institute of Protein Biochemistry, CNR, Naples. (B) Confocal microscopy image of a HeLa cell expressing a mitochondrial matrix RFP and an ER targeted GFP, in
which several appositions between the two organelles are visible (yellow spots). Scale bar: 10 ␮m. The inset highlights some specific points in which ER tubules seem to wrap
mitochondria (white arrows), likely marking sites for future mitochondrial fission (see text for details). (C) The cartoon schematically represents an ER-mitochondria contact
site, with some proteins proposed to play a structural role at this interface (see text for details). Traces on the right box represent simulated changes in [Ca2+ ] over time in
the bulk cytosol (black, dotted trace), in the mitochondrial matrix (red trace) and at regions of ER-mitochondria interface (black, continuous trace), upon an IP3-mediated
ER Ca2+ release. Note the different scales and the almost immediate generation of Ca2+ hot-spots on mitochondrial surface after IP3Rs opening. (For interpretation of the
references to colour in this figure legend, the reader is referred to the web version of this article.)

Fig. 2. Cell Functionalities relying on ER-mitochondria interactions. The cartoon summarizes the major reported functions modulated by ER-mitochondria juxtaposition,
and the principal molecules involved in these cross-talks, as described in the text or in other contributions (see for example [60,82]). For all the abbreviations used and the
description of specific functions, refer to the text. In italic, yeast proteins.

pletely elucidated and there is no direct evidence to suggest that endomembranes traffic and apoptosis [25]. For example, the PACS-
it acts as an ER-mitochondria tether: although depletion of the 2/BAP31 complex is involved in Bid translocation to mitochondria
protein causes mitochondria uncoupling from the ER, a BAP31- and apoptosis induction (Fig. 2), and a mitochondria-ER platform
dependent mitochondria fragmentation is also observed in these formed by the OMM protein Fis-1 and the ER resident protein BAP31
conditions [24], which could indirectly influence ER-mitochondria has been reported to be involved in the recruitment/activation of
association. Moreover, PACS-2 has a complex role in modulating pro-caspase 8 and in the subsequent cell death pathway [26]. Again,
4 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

whether Fis-1/BAP31 is just a functional complex or is endowed (jSR)-mitochondria close contacts length, but the number of con-
with an additional tethering capacity has not been established. tacts was not evaluated. However, given that in cardiomyocytes
The Familial Alzheimer’s Disease (FAD) related proteins the structures of both SR and mitochondria are highly ordered and
Presenilin-1 and −2 (PS1/2) have been also reported to be enriched continuously rearranged with age, making the jSR-mitochondria
in MAM [27]. However, only PS2, but not PS1, is able to favor physi- apposition a peculiar case significantly different from that of the
cal and functional ER-mitochondrial coupling, with FAD-linked PS2 ER-mitochondria connection (for a recent review, see [39]), a
mutants more effective than the wild type (wt) protein [28,29] (see direct comparison between the two structures should be avoided,
also below). Recently, it has been demonstrated that PS2 is able to especially considering the dramatic changes in mitochondrial and
increase the number of ER-mitochondria close contacts only in the potentially SR morphology induced by Mfn2 ablation.
presence of mitofusin 2 (Mfn2), a master regulator of organelles A remarkable exception is the elegant paper of Schneeberger
tethering whose action has been recently debated (see below): by et al. [40], where a careful quantification of ER-mitochondria
sequestering the latter protein and removing its negative effect juxtaposition was performed in POMC neurons. The authors
on organelles tethering, PS2 enhances ER-mitochondria association demonstrated that in obese mice there is a reduction in ER-
and Ca2+ transfer [30] (Fig. 1C). Whether PS2 represents one of the mitochondria contacts in POMC neurons and a reduction in Mfn2
possible multiple tethers between ER and mitochondria, or sim- expression. Moreover, they demonstrated that the selective Mfn2
ply works as a pure bait for the stumbling block Mfn2 (see below), ablation in POMC neurons induces obesity and, in 18 weeks old
remains to be clarified. mice, a reduction in ER-mitochondria coupling. It is, however, dif-
ficult to establish whether this decreased inter-organelle contact
2.2. The elusive role of Mfn2 is a direct consequence of Mfn2 ablation or a consequence of the
resulting obesity (see also below).
Among the different proteins proposed to have a role in the In the attempt to investigate the effects of Mfn2 on ER-
formation/modulation of ER-mitochondria tether structures, Mfn2 mitochondria tethering, a breaking point came from a quantitative
deserves a specific issue. electron microscopy (EM) approach, demonstrating that in
In mammalian cells, the OMM resident Mfn2, involved in Mfn2−/− MEFs (the same cell line used in [31]) the number of close
mitochondrial fusion process, has been originally proposed as an contacts between the two organelles (distance <20 nm), as well as
ER-mitochondria tether [31]. Indeed, part of the protein resides in the percentage of OMM engaged in the formation of contacts, is
ER membranes, particularly in MAM, and has been demonstrated by doubled, compared to wt MEFs [41]. Notably, Mfn2 re-expression
crossed co-immunoprecipitation experiments to physically inter- in Mfn2−/− cells rescued these parameters, suggesting that the
act with the OMM resident Mfn2, or with its homologue Mfn1. observed phenotypes were not due to clonal adaptation. However,
The observation, by confocal microscopy, that Mfn2−/− mouse in the same study, the reduced ER-mitochondria co-localization
embryonic fibroblasts (MEFs) display a reduced ER-mitochondria index, previously found in Mfn2−/− MEFs by a confocal microscopy
co-localization [31], as well as the previously reported role of Mfns analysis [31], was confirmed [41], casting doubts on the validity of
in tethering adjacent mitochondria during their fusion [32,33], ini- the latter method to quantitatively analyze inter-organelle apposi-
tially argued in favor of the proposed role of Mfn2 as a tether. tion.
Functionally, Mfn2−/− MEFs were demonstrated to have a reduced Recently, it has been confirmed that Mfn2 ablation, or its acute
mitochondrial Ca2+ uptake upon ER Ca2+ release, further suggest- down-regulation, increases ER-mitochondria coupling, reconciling
ing that the two organelles could be more distant and creating EM and confocal microscopy analysis [8]. In particular, it has been
consensus on the fact that Mfn2 connects them. Based on this demonstrated that the paradoxically lower ER-mitochondria co-
model, a number of studies used genetic manipulation of Mfn2 as localization, obtained by fluorescence microscopy, is due to the
a tool to modulate ER-mitochondria coupling, further reinforcing fact that Mfn2 ablation deeply affects mitochondrial morphol-
the thinking of this protein as a bona fide tether. However, many of ogy, fragmenting the organelles and, at the same time, swelling
these studies just assumed the original model and interpreted the them. By computational simulations, it has been shown that any
obtained results as its consequence, lacking a direct evaluation of mitochondrial swelling, even in the presence of a higher per-
the effects of Mfn2 modulation on inter-organelle association. For centage of OMM in contact with the ER, induces an apparent
instance, Mfn2 down-regulation was found to have an impact on and artifactual reduction in the classical Manders’ and Pearson’s
autophagosomes formation at ER-mitochondria contact sites [34], co-localization coefficients. The problem was tackled by consid-
but the effects of Mfn2 modulation on inter-organelle tethering was ering only the perimeter of mitochondria, instead of their entire
not directly investigated. Similarly, the E3 ubiquitin ligase MITOL area/volume: by this method, even by confocal microscopy, the
was found to modulate ER-mitochondria apposition, and this was percentage of mitochondrial perimeter in contact with the ER was
correlated to its effect on Mfn2 ubiquitination, but the direct effects found higher upon Mfn2 ablation/down-regulation, in accordance
of Mfn2 on organelles coupling were only evaluated by Mander’s with EM data. Regarding the reduced mitochondrial Ca2+ uptake
colocalization coefficient [35], a method that has been later demon- originally observed in Mfn2−/− MEFs upon ER Ca2+ release (a feature
strated to be misleading whenever marked changes in organelle’s interpreted as a consequence of a looser ER-mitochondria associa-
morphology occur (see below and [8]). Moreover, the effects of tion in these cells [31]), it has been found that this clone shows a
MITOL were not verified in Mfn2−/− cells, and thus they may, or ∼50% reduction in MCU expression level, compared to wt MEFs [8].
may not, be related to Mfn2. On the same line, Mfn2−/− MEFs were Thus, the reduced mitochondrial Ca2+ rises cannot be considered in
observed to form less starvation-induced autophagosomes, due to these cells a readout of a decreased organelles apposition. Indeed,
an impairment of lipid transfer from mitochondria [36]; also in this acute Mfn2 knock-down in wt MEFs, that does not change MCU
case, Mfn2 ablation effects on ER-mitochondria tethering were just expression, resulted in increased mitochondrial Ca2+ peaks, when-
assumed and not measured. Moreover, a more recent paper from ever an IP3-mediated ER Ca2+ release was induced, and increased
the same group showed a strong correlation between up-regulated sensitivity to Ca2+ -mediated apoptosis [8].
mitochondrial fusion processes and autophagy [37], potentially A higher ER-mitochondria tethering, measured by EM, was also
explaining the results they have previously obtained in Mfn2−/− associated with lower levels of Mfn2 and Mfn1 by another inde-
MEFs (where mitochondrial fusion is impaired). pendent group [42,43]. Interestingly, a different role for Mfn1 and
Another study [38] found that conditional Mfn2−/− mice show, Mfn2 in the modulation of mitochondria contacts with smooth
in the heart, a 30% reduction in junctional sarcoplasmic reticulum and rough ER, respectively, was proposed [42]. More extended
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 5

ER-mitochondria juxtapositions (thickness < 25 nm), but not a vari- induced by a rapamycin treatment, previously shown by EM to
ation in their number, were reported in HEK cells in which Mfn2 force ER-mitochondria contacts formation [46], should be similar
was acutely down-regulated and, functionally, an increased Ca2+ in the two conditions. In fact, morphological alterations of both
shuttling between the two organelles was observed [44]. Finally, ER and mitochondria (as occurring in Mfn2−/− cells [31]) have a
the Alzheimer’s disease related protein Presenilin-2, by binding dramatic impact on protein mobility within the two organelles,
and inhibiting the antagonistic role of Mfn2, has been shown to as previously demonstrated by FRAP experiments [31]. This effect
favor ER-mitochondria physical and functional coupling [30]. Thus, could potentially alter the rapamycin-induced reconstitution of the
accordingly to all these new data, Mfn2 can be still considered a artificial tether (FRETmax ), which is based on a zippering effect
master regulator of ER-mitochondria coupling, but with an antag- allowed by the free lateral diffusion within organelles’ membranes
onistic role. of the two modified GFP-variants forming the tether [46]. Secondly,
Very recently, another paper came out by the same authors the reported reduction in Mfn2−/− cells of the FRETratio , calcu-
that originally proposed the ER-mitochondria tether role for Mfn2 lated as (FRETmax -FRETbasal )/FRETbasal [45], means that in these cells
[45]. This latter claimed “definitive” evidence for the specific issue FRETbasal is closer to FRETmax . Thus, the presented data, showing
and argued against the published findings showing the contrary that Mfn2−/− cells display a lower FRETratio , indicate that in these
[8,41]. The decreased ER-mitochondria co-localization, originally cells there is an increased, and not a decreased, ER-mitochondria
observed by confocal microscopy in Mfn2−/− MEFs by the same tethering, as previously demonstrated [8,41].
authors [31], is here justified by EM data showing that, upon Mfn2 From the functional point of view as well, some data appear
deletion, ER and mitochondria are 1–3 nm more apart compared inconsistent. The very small difference reported in organelles dis-
to controls (while no difference in this parameter was found previ- tance (∼2 nm) would have a marginal effect (<1%) on the [Ca2+ ]
ously [8,41] in the same cell lines, wt and Mfn2−/− MEFs). Moreover, generated at the ER-mitochondria interface, as it can be calculated
upon Mfn2 depletion, a robust reduction in mitochondrial Ca2+ [47], thus hardly explaining the reported marked reduction in mito-
uptake, upon IP3-linked stimulations, was found, confirming their chondrial Ca2+ uptake, upon cell stimulation [45]. The latter results
previous results [31]. The latter effect was proposed to be indepen- would be more likely explained by a mitochondrial depolarization
dent from variations in MCU expression level. Finally, employing (reported in Mfn2−/− MEFs [48,49]) or by a lower MCU expression
a previously reported FRET-based ER-mitochondria artificial teth- in Mfn2 ablated cells, compared to controls [8]. Regarding this latter
ers [46], a reduced FRETratio in Mfn2−/− cells compared to controls result, it has been argued that it was due to different cell densities,
has been reported and interpreted as a proof of decreased inter- showing, however, that only in control cells MCU level decreases
organelle contacts [45]. at high density, but not in Mfn2−/− cells [45], thus not explaining
Several points are however debatable in this contribution and, the previous finding. On the contrary, in the same contribution,
although a quantitative EM analysis was employed to investigate changes in MCU complex expression levels were reported: rep-
the issue at higher resolution, the paper does not provide any con- resentative Western blots for MCU, MICU1 and MICU2 revealed
vincing new evidence in favor of the original hypothesis. First of major variations in the expression of these proteins in cells ablated
all, the key parameter, i.e., the number of contacts between the of Mfn2. Moreover, the experimental approach of permeabilized
organelles, that was previously found doubled in Mfn2−/− cells cells perfused with different media containing fixed [Ca2+ ], to study
[8,41] was not measured. Instead, only ER-mitochondria inter- the intrinsic capability of mitochondria to take up Ca2+ , was here
face length, i.e., the averaged extension of each single contact, is criticized. It has been stated that results obtained by this protocol
reported to be decreased in Mfn2−/− cells (from 253 nm in wt to (showing that in Mfn2−/− cells there is a decreased mitochondrial
195 nm in Mfn2−/− cells; a feature not observed in previous studies Ca2+ uptake independently from the ER-mitochondria association
[8,41]). However, the quantification of this parameter is not enough [8]) are difficult to interpret because the presence of the ER, with its
to sustain a reduced ER-mitochondria juxtaposition in Mfn2−/− high Ca2+ affinity SERCA, could “contaminate” the outcome [45]. To
cells: if the contacts in these cells are more frequent (as it has been investigate the same issue, isolated mitochondria have been used
previously observed), the percentage of mitochondrial perimeter in instead. However, it must be noted that the classical permeabi-
contact with the ER could be increased. Thus, it is critical to exactly lization protocol, applied in multiple occasions by several groups
know the number of contacts, in addition to their extension, to (1 min in 100 ␮M digitonin-containing medium without ATP, fol-
solve this debated point. Furthermore, the reduced interface length, lowed by other 2 min washing with an intracellular-like medium
observed only in this study, cannot be chosen as an unequivocal without digitonin and ATP; [8]), does not allow any SERCA activ-
proof of ER-mitochondria apposition since the same paper showed ity. Instead, the presented protocol, isolating mitochondria from
that: Mfn1−/− cells (which present an unchanged ER-mitochondria mice liver without any specific purification step [45], leads to a
tethering [31]) display a similar reduction in this parameter, and mitochondrial fraction highly contaminated by ER vesicles.
Mfn2 re-expression in Mfn2−/− cells does not recover it, thus inval- Finally, Ca2+ data obtained in intact cells are also puzzling: it
idating also the bizarre ERMICC coefficient used [45]. Importantly, has been claimed that, upon cell stimulation, Mfn2 ablated cells
the difference in ER-mitochondria distance, found by EM analysis, exhibit a lower mitochondrial Ca2+ transient, compared to controls
between controls and Mfn2−/− cells (+1–3 nm in these latter cells) (as a proof of a reduced ER-mitochondria juxtaposition). However,
does not explain the substantial apparent reduction in organelles’ the presented data showed that the mitochondrial [Ca2+ ] peaks
co-localization, as previously observed by confocal microscopy obtained in controls, under a specific condition, can be variable till
[8,31,41]. Indeed, differences in this range of values are incompat- two order of magnitude [45], making any comparison with those of
ible with the resolution of confocal microscopy (at best ∼200 nm, Mfn2−/− cells very difficult. Moreover, upon MCU overexpression
i.e., 100 fold lower), and does not produce any outcome observable in wt and Mfn2−/− MEF cells, the reported mitochondrial [Ca2+ ]
by this technique. peaks, obtained upon cell stimulation, were very similar to those
Moreover, the interpretation of the FRET data reported for previously found by the same authors in the very same cells with
Mfn2−/− cells, and presented as an additional proof of a reduced endogenous levels of MCU [31]: given the well-known dramatic
ER-mitochondria juxtaposition, is questionable and argue against impact of the relative amounts of each MCU-complex component
the proposed conclusion. Firstly, raw values of basal and maximal on the process of mitochondrial Ca2+ uptake [50], these results
FRET are missing. These parameters are indeed crucial: to allow, by appear difficult to explain.
this method, an indirect comparative quantification of organelle Thus, since this latter contribution does not significantly modify
contacts in wt and Mfn2−/− cells, maximal FRET values, artificially previous multiple conclusions that sustain a negative role of Mfn2
6 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

in ER-mitochondria tethering [8,30,41–44], the specific mechanism membrane protein PTPIP51, have been reported to be involved in PS
by which this protein modulates organelles’ coupling remains elu- transfer [22] (Fig. 2). As far as mitochondrial sterols import is con-
sive. Likely, Mfn2 physically interacts with and inhibits the proper cerned, recent studies have provided the first possible molecular
assembly of still unknown tethering proteins (Figure 1C), though identity in the OMM translocator protein (TSPO) [67]. TSPO seems
further investigations will be necessary to address this issue. to be enriched in MAM by interacting with VDAC [68] (Fig. 2).
ER-mitochondria connections play also a key role in controlling
the morphology and the dynamics of the mitochondrial network
3. Functions: cellular pathways dependent on (Fig. 2). Indeed, mitochondrial fission occurs at positions where
ER-mitochondria coupling ER contacts mitochondria [69], mediating organelle constriction
before the recruitment of dynamin-related protein 1 (DRP1), form-
3.1. Lipid homeostasis, mitochondria dynamics, autophagy, ing helical oligomers that wrap around the organelles and divide
inflammation and mtDNA distribution them in a GTP-dependent manner [70]. In metazoan cells, mito-
chondrial division mediated by ER constrictions involves also actin
ER-mitochondria contacts, in both yeast and mammals, repre- polymerization through the ER protein inverted formin-2 (INF2),
sent important structures linked to multiple pathways, ranging a promoter of actin polymerization/depolarization [71,72]. Mito-
from different mitochondria functionalities to several cell pro- chondrial fission is an essential event involved in mitochondrial
cesses, such as proliferation, death, autophagy, lipid metabolism, network shaping and is required to generate organelles that are
Ca2+ signaling, bioenergetics, unfolded protein response (UPR) at the right size to be transported by molecular motors along the
and inflammation [4]. While for some of them the role of ER- cytoskeleton. Moreover, mitochondrial fission is important for the
mitochondria coupling has been extensively studied (for example, release of cytochrome c and other pro-apoptotic factors from the
on lipid synthesis, Ca2+ homeostasis, mitochondria metabolism mitochondrial inter-membrane space to trigger cell death, and
and cell death; see below), for others, evidence has started to it is thought to facilitate the removal of damaged organelles by
accumulate only very recently. Thus, the ER-mitochondria axis is mitophagy [73].
progressively emerging as a complex signaling platform fundamen- Moreover, it has been recently shown that autophagosomes
tal for cell survival. can originate at ER-mitochondria contact sites. Key components
The first cellular function demonstrated to rely on ER- of the autophagy process (Atg5, Atg14, Beclin-1, Vps15, Vps34)
mitochondria juxtaposition was the maintenance of membrane were found in MAM under starved conditions (and this localization
lipid composition. Indeed, lipid synthesis and transfer are the more was stable throughout all the autophagosome formation process)
prominent features of ER-mitochondria contact domains: MAM and alterations in ER-mitochondria tethering led to decreased
were originally defined by Jean Vance as fraction X, biochemi- autophagy [34]. In addition, fundamental processes, such as lipida-
cally distinct from the bulk ER membranes because particularly tion of LC3-I into LC3-II and the recruitment of calnexin to raft-like
enriched in enzymes catalyzing phosphatidylserine (PS), phos- microdomains containing glanglioside-3 (GD3) [74,75], have been
phatidylethanolamine (PE) and phosphatidylcholine (PC) synthesis shown to depend on ER-mitochondria connection (Fig. 2), placing
[51,52]. In fact, although the ER is the master organelle for lipid syn- this inter-organelle interface at the heart of the autophagy pro-
thesis, certain phospholipids synthetic pathways require enzymes cess. Very recently, the OMM protein FUNDC1, which functions
that are located in both the ER and the mitochondria: for exam- as LC3 receptor during hypoxia-induced mitophagy [76], has been
ple, the biosynthesis cycle of PC and PE starts from PS synthesis at also found to be enriched in MAM and function as an adaptor
ER-MAM domains by PS synthase [53,54], an then follows its decar- protein coordinating mitochondrial dynamics and quality control,
boxylation into PE at the inner mitochondrial membrane (IMM), in response to different signaling pathways and mitochondrial
and back to the ER for conversion into PC [51,55,56]. Linked to their stresses. Depending on its phosphorylation pattern, FUNDC1 can
synthesis, there is also an extensive lipid exchange between ER interact with calnexin, DRP1 and OPA1, mediating either mito-
and mitochondria membranes: PC is the most abundant mitochon- chondria elongation in response to hypoxic stimuli or mitochondria
drial phospholipid and, together with PS, must be imported from fragmentation and mitophagy [77,78].
the ER through yet uncharacterised mechanisms, likely involv- A further cell process that could involve ER-mitochondria
ing ER membranes and OMM close juxtapositions [57]. Moreover, contacts is the formation and regulation of inflammasomes, multi-
ceramides synthesis has been shown to occur at MAM [58] and molecular complexes that work as a platform for the processing
these latter have been also described as microdomains enriched in and release of pro-inflammatory cytokines [79]. One of these com-
cholesterol and gangliosides, modified sphingolipids, similarly to plexes, the Nod-like receptor NLRP3, that mediates the activation
the detergent-resistant lipid rafts at the plasma membrane [59]. of interleukins IL1␤ and IL18, localizes, when inactive, to the ER but,
The molecular mechanisms allowing ER-mitochondria lipid upon inflammosome activation, redistributes to ER-mitochondria
exchange (Fig. 2) are mostly unknown [60] and insights come clusters comprising MAM [80]. Since one of the physiological trig-
almost exclusively from yeast: both the ERMES and the ECM com- gers of NLRP3 activation is ROS, and given that mitochondria and
plex are involved in lipid trafficking between the organelles [61,62]; ER are major intracellular locations for ROS production, MAM
ERMES has Synaptotagmin-like Mitochondrial lipid-binding Pro- represent a privileged sites enriched in ROS [81] where the inflam-
tein (SMP) domains [63] that favor the transfer of lipids (especially masome becomes active (Fig. 2). Interestingly, upon knock-down
PC) through tubular, hydrophobic structures [64]. Some yeast of VDAC1, which is enriched in MAM and mediates mitochon-
members of the oxysterol-binding homology (Osh) proteins fam- drial Ca2+ entry (see below), inflammasome activation is reduced
ily are also necessary for proper PS homeostasis [13]. Recently, [80], indicating that ER-mitochondria Ca2+ cross-talk and conse-
it has been suggested that sterols are directly transported from quent mitochondrial ROS production play a crucial role in this
the ER to the mitochondria through Ltc1/Lam6, an ER-located phenomenon (see [82–84] for recent reviews). Moreover, in cystic
sterol transporter enriched in MAM [16] and interacting with the fibrosis cell models, it has been recently shown that mitochon-
ERMES complex [65,66]. Ltc1/Lam6 also likely functions as a tether drial Ca2+ uptake integrates pro-inflammatory signals initiated
between ER and mitochondria by interacting with the OMM pro- by pathogen-associated molecules and relays the information to
teins Tom71/70 [65]. NLRP3 [85], acting as a synergic stress stimulus in triggering exac-
In mammalian cells, ORP5 and ORP8, which are present at ER- erbated inflammatory response.
mitochondria contact sites and interact with the mitochondrial
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 7

Physical ER-mitochondria juxtapositions have been recently cycle, are modulated by matrix Ca2+ [103]. The first one (Pyru-
reported to be spatially linked to mtDNA synthesis and distribution vate Dehydrogenase, PDH) converts pyruvate into acetyl-CoA, that
in dividing mitochondria in both yeast [60,86] and human cells [87], enters into the Krebs cycle. PDH is active when de-phosphorylated
supporting the idea that ER-mitochondria contacts are involved in and Ca2+ activates its phosphatase. The other two enzymes (Isoc-
the distribution of mtDNA copies to daughter mitochondria during itrate Dehydrogenase, ICDH, and Oxoglutarate Dehydrogenase,
the division process. OGDH) are both regulated by Ca2+ through its direct binding and
allosteric modulation (Kd of 20–30 ␮M and 1 ␮M, respectively
3.2. ER-mitochondria Ca2+ cross-talk functionalities [104]), inducing an increased affinity for their substrates. A fourth
Ca2+ -sensitive mitochondrial dehydrogenase, the FAD-dependent
Ca2+ uptake into mitochondrial matrix is known to occur since Glycerol-phosphate Dehydrogenase (GPDH), is located in the IMM
the pivotal studies on isolated mitochondria in the 60 s [88]. It was and senses the cation concentration in the intermembrane space
early clear that the electrochemical gradient (, ∼ -180 mV, neg- (IMS), thanks to a here located Ca2+ binding motif (Kd for Ca2+
ative on the side of the matrix), generated by the activity of the of 0.1 ␮M). GPDH transfers reducing equivalents from glycolytic
respiratory chain across the IMM [89], represents the ultimate ther- NADH to the electron transport chain (ETC) as FADH2 and favors
modynamic basis responsible for the import of the cation. However, ATP production. Moreover, it has been reported in isolated mito-
in the 70 s, the accurate definition of the characteristics/kinetics chondria that Ca2+ can directly stimulate the ETC and the F0 F1
of the specialized transport mechanisms that strictly control the ATP synthase activity [105,106]. In addition to the above described
process [90], whose molecular identity has been recently clarified matrix Ca2+ effects, specific IMM-located mitochondrial carriers
[91,92], revealed that the so called “Mitochondrial Ca2+ uniporter” for different nucleotides/metabolites/co-factors are modulated by
(MCU) is activated and allows an appreciable uptake of the cation IMS [Ca2+ ]. These Ca2+ -binding mitochondrial carriers (CaMCs) are
into the matrix only in the presence of relatively high [Ca2+ ]. usually endowed with N-terminal EF Ca2+ -binding domains facing
The interested readers are referred to some recent reviews for an the IMS [107,108]. Recently, in a very elegant study, a constitutive,
overview on MCU complex and other transporters identity/activity low-level IP3-mediated ER Ca2+ release has been demonstrated to
[93–95]. be essential for the stimulation of mitochondrial ATP production,
In particular, the low MCU affinity for Ca2+ (Kd ∼ 15–20 ␮M) likely through mitochondrial uptake of the released ER Ca2+ and
does not match the physiological [Ca2+ ] reached in the cytosol the above described stimulatory effects [109]. Indeed, the block of
of living cells, ranging from 50 to 100 nM in resting conditions this ER-mitochondria Ca2+ shuttling (through the IP3Rs inhibitor
to peaks of 1–3 ␮M, upon stimulation. In the early 90 s, the find- Xestospongin B or by genetic IP3Rs deletion) deeply impaired
ing that, upon cell stimulation, mitochondria take up Ca2+ with an mitochondrial ATP synthesis, leading to an increased intracellular
amplitude much higher than the predicted one was thus initially AMP/ATP ratio, AMPK activation and a potent autophagy induction.
puzzling [96]. These surprising results were obtained in living cells Similarly, stable MCU/MCUR1 knockdown, that dampens mito-
thanks to the introduction of genetically encoded Ca2+ indicators chondrial Ca2+ uptake, reduces cellular oxygen consumption rate,
(firstly aequorin [96]) specifically targeted to mitochondrial matrix activates AMPK and induces autophagy [110]. The pro-survival
(see [97] for a recent review on different mitochondria-targeted effect of a physiological ER-mitochondria Ca2+ transfer has been
Ca2+ probes). The puzzle was solved by the observation that part also suggested by a transient increased ER-mitochondria coupling
of mitochondria is strategically located in very close apposition during the early phases of ER-stress and unfolded protein response
to the sites of Ca2+ release from the ER [2,98] (see also above). (UPR) [111]. This strengthened physical apposition has been inter-
In these regions, the fast opening of specific Ca2+ channels on ER preted as an attempt to overcome ER stress, energizing the cells
membranes (such as IP3Rs and/or RyRs) induces the generation by stimulating mitochondrial ATP production via increased matrix
of high [Ca2+ ] microdomains in proximity of their mouths. Juxta- [Ca2+ ] (Fig. 2).
posed mitochondria experience these Ca2+ hot-spots (i.e., they are While physiological mitochondrial [Ca2+ ] stimulate ATP pro-
locally exposed to a [Ca2+ ] one order of magnitude higher than that duction, excessive accumulation of the cation in the matrix (Ca2+
of the bulk cytoplasm [46,99]), allowing to overcome the thresh- overload) leads to the so called “permeability transition”. This
old for a prompt MCU activation and a fast mitochondrial Ca2+ latter, mediated by a Ca2+ -sensitive, high-conductance and voltage-
uptake (Fig. 1C). On the other hand, the latter process locally buffers dependent channel (mPTP) represents an increased permeability of
cytosolic Ca2+ rises, deeply affecting the allosteric modulation of the IMM to small molecules (up to 1500 Da), that in turn induces
ER Ca2+ -releasing channels. The details of the process that leads mitochondrial depolarization, impairs oxidative phosphorylation,
to the generation of Ca2+ microdomains and their relationship to increases ROS production, leads to mitochondrial swelling and
mitochondria has been recently reviewed [39], as well as the role OMM rupture, with release of IMS-resident pro-apoptotic factors
and significance of mitochondrial Ca2+ uptake in cell functionality (reviewed in [112,113]). Among them, of particular importance are
[100]. This contribution will be limited to a brief summary of the cytochrome c, apoptosis induction factor (AIF), Smac/DIABLO, pro-
main aspects of mitochondrial functionality regulated by Ca2+ , tak- caspase 9 and endonuclease G. The release of these factors induces
ing into particular consideration the signaling events that are more three different events (reviewed in [113]). The first is the assem-
directly affected by the close proximity with the ER. bly in the cytosol of the “apoptosome” (induced by cytochrome c
On the consequences linked to [Ca2+ ] rises within mitochon- interaction with the apoptosis protease activating factor 1, APAF1),
dria, two possible and opposite effects can be achieved: stimulation which activates pro-caspase 9 and effector caspases (caspase-3, −6,
of the Krebs cycle, NADH formation and fueling of the respira- −7). The second is the inhibition, by Smac/DIABLO, of caspase’s
tory chain activity, with increased ATP production; or sensitization inhibitors, such as inhibitor of apoptosis protein (IAP). The third is
towards the intrinsic apoptosis pathway, due to mitochondrial Ca2+ represented by DNA fragmentation and chromatin condensation,
overload, opening of the mitochondrial Permeability Transition by AIF and endonuclease G.
Pore (mPTP) and release of cytochrome c (Fig. 2). The molecular identity of the mPTP remained elusive up to
As to the former, it is well established that transient increases few years ago. It was firstly demonstrated that the c subunit of
in matrix [Ca2+ ], such as those obtained upon an IP3-dependent the ATP synthase is a major regulator of mPTP activity [114];
ER Ca2+ release, stimulate NADH production and ATP synthe- then, dimers of the F0 F1 ATP synthase have been proposed to
sis [101,102]. Indeed, three different enzymes, one immediately form the mPTP [115]. Though there is consensus on the fact
upstream of, and the other two directly taking part in the Krebs
8 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

that the ATP synthase is an essential component of the mPTP, 4. Significance: dysfunctions in ER-mitochondria coupling
more debated is the exact definition of which ATP synthase sub- and diseases
units directly take part in its formation [116]. Moreover, recently,
a different mechanism was proposed to mediate mitochondrial Since communication between ER and mitochondria regulates
Ca2+ -induced cell death, represented by the Ca2+ binding to the a multitude of physiological processes (see above), it is not surpris-
IMM-located cardiolipin, cardiolipin coalescence, disassembling of ing that recently alterations in organelles’ physical and functional
the cardiolipin-associated respiratory complex II, release of a ROS tethering have been proposed as a common hallmark in differ-
generating sub-complex and cell death triggering [117]. Finally, the ent pathologies, such as neurodegenerative diseases, metabolism
importance of the ER-mitochondria platform in determining cell syndrome and cancer. A comprehensive and detailed dissertation
life or death decisions is further strengthened by multiple evidence on this aspect is far away from the scope of the present contribu-
indicating that different oncogenes and tumor suppressors actively tion. We here present a brief summary of the main studies finding
shape ER to mitochondria Ca2+ transfer (see below and [118,119]). alterations in ER-mitochondria pathways that can be linked to the
ER-mitochondria coupling is also crucial in modulating cell pathogenesis of specific human diseases (Table 1). The interested
metabolism. Indeed, at cellular level, metabolism relies on differ- reader is also referred to additional reviews on this topic [127–129].
ent complementary and exclusive pathways, compartmentalized
within the eukaryotic cell. In this regard, the regulation of metabolic 4.1. Metabolic syndrome and inflexibility
pathways, in order to maintain proper cell energy homeostasis,
depends on the activity of different organelles. In particular, ER Mitochondria and ER activity are essential in the control of
and mitochondria are two major organelles controlling cellular metabolic homeostasis (see above) and, in general, impaired func-
metabolism and energy production. On one hand, mitochondria tions of the ER [130] and mitochondria [131] have been associated
are the conductor in adapting energy production to energy require- with obesity, insulin resistance (IR) and metabolic defects. Given
ments. By hosting the Krebs cycle, which is the end-point of lipid, the inter-dependency of these two organelles, the ER-mitochondria
glucose and glutamine catabolism, as well as the ETC responsible axis has recently emerged as a potential central actor in the
for the production of ATP through the consumption of reductive pathophysiology of metabolic syndrome [132]. Indeed, genetically
power, mitochondria have a major impact on metabolite fluxes, or diet-induced obesity and IR have been associated with dis-
energy charge and redox state of the cell. On the other hand, criti- rupted ER-mitochondria interactions in liver and neurons [40],
cal steps of glucose and lipid anabolism take place within the ER, as even though a debate remains whether organelles’ juxtaposition
well as protein synthesis and sorting, making the organelle a cru- is increased [133] or decreased [126,134] in models of hepatic IR.
cial actor in metabolic homeostasis. Hence, proper cell metabolism However, growing evidence indicates that experimental disrup-
is directly linked to the proper activity of these two compartments, tion of ER-mitochondria interactions can induce IR. For example,
which in turn is relying on their mutual interaction. the knock-out of a subunit of mTORC2, which induces a reduced
As described above, Ca2+ transfer from the ER to mitochondria ER-mitochondria association in MEF cells [124], has been associ-
stimulates, and is necessary for, mitochondrial activity, regulat- ated with IR in the liver [135]. Moreover, Mfn2 ablation (to alter,
ing cell redox state and ATP production [101,102]. However, the in a positive or negative manner (see above), the physiological ER-
importance of Ca2+ homeostasis in the control of metabolism is not mitochondria interaction) has been associated in POMC neurons
limited to mitochondria. Indeed, cytosolic Ca2+ directly activates with leptin-resistance, leading to obesity [40] (but see also above),
glucose metabolism by stimulating the activity of gluconeogene- and with IR in hepatocytes and myotubes [136].
sis and glycogenolysis enzymes in the liver [120]. Moreover, the Ca2+ is likely to represent the mechanistic link between
action of glucagon and catecholamines, hormones participating ER-mitochondria interaction and metabolic impairment. Indeed,
in the control of glucose metabolism, is mediated by increases in disruption of Ca2+ homeostasis has been shown to mediate the
cytosolic Ca2+ and subsequent positive modulation of calmodulin- suppression of hepatic insulin signaling through Ca2+ /calmodulin-
dependent kinases and calcineurin, leading to the activation of dependent protein kinase II hyper-activation [137] and to link
the transcription factor FOXO [121]. In this pathway, ER-located mitochondrial dysfunction to ER stress and hepatic IR [138]; the
IP3Rs play a central role, mediating cytosolic Ca2+ oscillations, later phenomenon was in fact counteracted by SERCA overex-
through the release of the cation from the ER but also through pression to restore ER Ca2+ homeostasis in the liver of obese
its direct interaction with the cAMP response element-binding mice [139]. Moreover, specific alteration of mitochondrial Ca2+
protein-regulated transcription coactivator 2 (CRTC2) promoting uptake can disrupt insulin signaling in cardiomyocytes [140],
its calcineurin-mediated phosphorylation/activation [122]. Thus, and IP3R1 heterozygous mutant mice develop glucose intoler-
the importance of the ER-mitochondria axis in the control of ance and hypersensitivity to diet-induced metabolic syndrome
cellular Ca2+ homeostasis gives to this interface a major role [141]. Likewise, mice knock-out for CypD, a protein that modulates
in the control of metabolism, not only by stimulating mito- ER-mitochondria coupling in liver and cardiomyocytes [126,142],
chondrial activity but also regulating the action of different have been shown to develop hepatic IR through MAM impair-
hormones. Interestingly, insulin, a major actor in the control ment and subsequent disruption of Ca2+ homeostasis [143], while
of the entire body metabolism, induces an inhibitory phospho- ablation of the MAM-located protein cisd2 induces cytosolic Ca2+
rylation of IP3Rs by activating the MAM-located enzyme AKT de-regulation, mitochondrial dysfunction and IR in the adipose tis-
[123]. Moreover, mTORC2 and PTEN, two proteins which are sue [144]. If these studies provide convincing proofs that MAM
part of the insulin signaling cascade, are also present at the impairment can lead to disrupted metabolism, it is likely than an
ER-mitochondria interface [124,125], and mTORC2 and phospho- excess of ER-mitochondria communication can have similar con-
rylated AKT are recruited to MAM upon insulin stimulation in the sequences, as shown by the fact that liver specific overexpression
liver [124,126]. of PACS-2 or IP3R1 in mice is linked to IR [133].
By this dual role in Ca2+ -mediated control of mitochondrial On this line, tools to modulate ER-mitochondria interactions
activity, cytosolic enzymes and insulin signaling, ER-mitochondria could represent an efficient approach to restore metabolic homeo-
platform appears as a crucial element in the maintenance and stasis. Indeed, hepatic insulin sensitivity can be rescued in models
adaptation of cell metabolism to external stimuli and energy of obesity and IR, in which MAM have been shown to be decreased,
requirements. through the overexpression of CypD [126]. An interesting ele-
ment is that there seems to be an interdependency between
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 9

Table 1
Main disease-related proteins, and relative interactors, reported to modulate ER-mitochondria coupling. See text for details.

PROTEINS FUNCTIONS INTERACTORS ASSOCIATED DISEASES REFERENCES

VAPB Physically/functionally tethers ER to mitochondria by PTPIP51 Amyotrophic Lateral 19; 20


interacting with the OMM-located PTPIP51 Sclerosis (ALS)
PTPIP51 Physically/functionally tethers mitochondria to ER by VAPB ALS 19; 20
interacting with the ER-located VAPB
TDP-43 Disrupts ER-mitochondria apposition by a GSK-3␤-mediated GSK-3␤ ALS 20
disassembly of the VAPB-PTPIP51 complex (VAPB/PTPIP51)
FUS Disrupts ER-mitochondria apposition by a GSK-3␤-mediated GSK-3␤ ALS 21
disassembly of the VAPB-PTPIP51 complex (VAPB/PTPIP51)
SOD1 Expression of hSOD1-G93A mutant alters ER-mitochondria ? ALS 183
Ca2+ homeostasis
SIGMA1-R Loss of SIGMA1-R or expression of loss-of-function mutants IP3Rs ALS; Distal Hereditary 184; 185
impair ER-mitochondria communication Motor Neuropathies
(dHMNs)
PS2 Expression of PS2, especially of FAD-mutants, favors Mfn2 Alzheimer’s disease 28; 29; 30
ER-mitochondria physical/functional coupling, by sequestering (AD)
Mfn2
Mfn2 Modulates either positively or negatively ER-mitochondria Mfn2/Mfn1; PS2; Charcot-Marie-Tooth 31; 35; 41; 8
physical/functional juxtaposition MITOL Neuropathy Type 2A 40; 136
(CMT2A)
Obesity, Insulin
resistance (IR)
AKT/PKB Inhibits ER Ca2+ release and negatively affects Ca2+ -dependent IP3Rs Cancer 123; 155; 156
apoptotis by phosphorylating IP3Rs
PTEN Sensitizes cells to Ca2+ -dependent apoptosis by IP3Rs Cancer 125
de-phosphorylating IP3Rs and promoting its activity
PML Promotes ER-mitochondria Ca2+ transfer and apoptosis by IP3R3/AKT/PP2A Cancer 157
forming a complex containing IP3R3, AKT and PP2A. PP2A
suppresses AKT-mediated IP3R3 phosphorylation, favoring its
activity
BRCA1 It is recruited to ER during apoptosis, sensitizing the IP3Rs to IP3Rs Cancer 158
low [IP3]
FATE1 Uncouples ER from mitochondria, protecting from Ca2+ - and ? Cancer 159
drug-induced cell death
PARKIN Favors ER-mitochondria coupling, increasing mitochondrial ? Parkinson’s disease 174
Ca2+ uptake and sustaining ATP synthesis (PD)
␣-synuclein Favors mitochondrial Ca2+ uptake by enhancing ? PD 177; 178
ER-mitochondria apposition; disease-associated mutants
uncouple the two organelles
DJ-1 Promotes ER-mitochondria coupling by counteracting p53 p53 PD 179; 180
activity; wt, but not pathogenic mutants, sustains ATP
production
mTORC2 Promotes ER-mitochondria coupling and has been proposed to IP3R-Grp75-VDAC1 IR 124; 135
be an MAM signaling hub by controlling AKT mediated complex
phosphorylation of IP3R, HK2 and PACS-2 AKT
PACS-2
HK2
IP3R1 ER Ca2+ release channel Grp75 IR 133; 141; 18;
VDAC1 Metabolic syndrome 123; 125
AKT
PTEN
PACS-2 Favors physical/functional ER-mitochondria juxtaposition and Bid IR 24; 26; 133
controls Bid-mediated apotosis Fis-1/BAP31
CypD Promotes ER-mitochondria coupling through an unknown IP3R-Grp75-VDAC1 IR 126; 142; 143
mechanism complex Hypoxia-
reoxygenation
injury
cisd2 Although its function remains unclear, this protein is crucial in ? Wolfram syndrome 144
the maintenance of Ca2+ homeostasis and ER/mitochondria IR
function

ER-mitochondria coupling and metabolic homeostasis since high- increasing and decreasing organelles coupling lead to metabolic
fat diet and CypD knock-out mice, two models in which hepatic dysfunctionalities can be re-conciliated within the idea that for cell
IR have been linked to reduced interactions between ER and metabolism, the strength of ER-mitochondria coupling are neither
mitochondria, exhibited a restored MAM quantity after insulin sen- good nor bad. The right amount needs to be suited to the cellu-
sitivity improvement upon specific pharmacological treatments lar state, and both Ca2+ overload and impaired Ca2+ uptake could
[126]. However, these findings are still under intensive debate, lead to mitochondrial dysfunctions. However, the understanding of
since a major study showed opposite results, i.e., in the liver of a why similar experimental models and tissues can produce opposite
genetic mouse model of obesity and IR (ob/ob mice), an increased outcomes modulating the MAM state will require further investi-
interaction between the two organelles, leading to Ca2+ -mediated gation. Part of the answer probably resides in the highly dynamic
mitochondrial dysfunction, and an ameliorated insulin sensitivity, characteristic of both metabolism and ER-mitochondria interac-
upon inhibition of organelle juxtaposition by the knocking-down tions, in which adaptive responses to metabolic challenges and
of PACS-2 or IP3R1, have been observed [133]. The fact that both environmental stimuli can influence MAM formation/stability.
10 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

IR is usually seen as a disruption of a cellular signaling pathway. fraction of PTEN, that increases after apoptosis stimulation with
A more recent and complete way of approaching metabolic dis- arachidonic acid: the resulting increased ER Ca2+ release induces
eases is to picture metabolism as a flexible network of enzymes mitochondrial Ca2+ overload, sensitizing cells to apoptosis [125].
and metabolites that adapts to external nutritional stimuli, a The tumor suppressor PML has been shown to act on the same path-
phenomenon called metabolic flexibility [145]. Thus, metabolic way as well: by interacting with IP3Rs, it promotes the formation
impairment can be seen as the inability of the organism to switch of a complex containing IP3R3, AKT and the protein phosphatase
nutrient sources between fatty acid and glucose in different ener- PP2A. In turn, PP2A suppresses AKT-mediated IP3R3 phosphoryla-
getic contexts: fasting and fed states, physical activity or hormone tion, thus promoting ER-mitochondria Ca2+ transfer and apoptosis
stimulation [145]. Given their importance in energy homeostasis, [157]. The tumor suppressor proteins BRCA1 was found to be
ER and mitochondrial functions are crucial in adapting metabolism recruited to the ER during apoptosis in an IP3R-dependent manner,
to nutrient availability and have been shown to be key actors in sensitizing the IP3R to low [IP3], thus favoring the Ca2+ -dependent
metabolic flexibility at the cellular level [146]. apoptotic response [158]. Recently, the cancer-testis antigen FATE1
ER-mitochondria coupling has recently emerged as a new actor has been reported to uncouple ER from mitochondria, protect-
in the modulation of metabolism. Indeed, the adaptation of this ing from Ca2+ - and drug-induced cell death [159]. Though cancer
functional relationship participates in the control of mitochon- cells avoid massive mitochondrial Ca2+ uptake, as a mechanism
drial activity regarding energy requirements and metabolic context to escape from Ca2+ -overload and apoptosis, a constitutive, low
[134]. In particular, MAM quantity increases during fasting in the level ER to mitochondria Ca2+ transfer is essential for their viabil-
liver [147], a phenomenon that was shown to be controlled by ity. Indeed, very recently, it has been demonstrated that a basal
the modulation of the pentose phosphate-protein phosphatase mitochondrial Ca2+ uptake is necessary to sustain tumor cells
2 A (PP2A) pathway by glucose availability, leading to subsequent bioenergetics and to provide them with essential metabolites [160].
mitochondrial fusion and increased Ca2+ uptake and metabolism Overall, these recent findings highlight the importance of the mod-
of the organelle [148]. Interestingly, mitochondrial dysfunction ulation of ER-mitochondria interface in cancer cells.
has been implicated in metabolic inflexibility [146], and a recent
study demonstrated an impairment of ER-mitochondria coupling 4.3. Alzheimer’s disease
modulation upon fasting in the liver of IR mice, associated with
mitochondrial fragmentation and decreased oxygen consumption Alzheimer’s disease (AD) is an irreversible neurodegenerative
[148]. This observation is supported by the finding that a chronic disorder, whose pathogenesis has been classically considered as
hyper-activation of PP2A, a negative pathway for MAM formation, strictly dependent on an altered production of the amyloid beta
has been found in the IR liver [149,150]. (A␤) peptide. The idea that A␤ generation is the key event, whose
perturbation is at the basis of AD onset, found strong genetic
4.2. Cancer supports in the middle of the 90’s, with the discovery that the
familial forms of AD (FAD) are linked to autosomal dominant muta-
Over the last years, increasing evidence has been provided on tions in three genes, encoding for amyloid precursor protein (APP),
the capacity of cancer cells to remodel intracellular Ca2+ signaling, Presenilin-1 (PS1) and Presenilin-2 (PS2). Indeed, A␤ is generated
favoring their survival and proliferation. Among multiple mech- from APP, thanks to its cleavage by the ␤- and the ␥-secretases;
anisms, enhanced resistance to apoptosis of cancer cells involves PS1 and PS2 (as alternative catalytic core of ␥-secretase) are fun-
modulation of the ER-mitochondria Ca2+ cross-talk. Several tumors damental in the process. Though the pathogenicity of A␤ has
display an altered expression of proteins, including oncogenes and been well established, so far many efforts aimed at controlling the
tumor suppressors, that directly affect MAM functionality and in generation or the accumulation of the different species of the pep-
particular ER-mitochondria Ca2+ transfer [151,152] (Fig. 2). Below, tide (typically, A␤40 and A␤42) failed to convincingly impact on
we provide few examples, and the interested readers are referred the onset/progression of the pathology, suggesting that additional
to a recent review for a more complete list of the proteins that have molecular mechanisms may be altered since the early phases of the
been involved in these pathways [119]. disease. In addition to anomalies in A␤ generation, AD patients dis-
In general, tumor suppressors enhance ER-mitochondria pro- play different early intracellular alterations, such as Ca2+ and lipids
apoptotic Ca2+ transfer, while oncogenes dampen it. For instance, dyshomeostasis, increased ROS levels, impairments in autophagy,
the oncogenes Mcl-1, Bcl-2 and Bcl-XL decrease ER Ca2+ con- axonal transport and mitochondrial dynamics. Interestingly, MAM
tent, likely by increasing the basal IP3R-dependent Ca2+ leak, are the central hub where many of these activities are controlled or
avoiding ER-derived mitochondrial Ca2+ overload upon stress con- directly take place. Thus, very recently, the hypothesis that AD may
ditions (reviewed in [119]). On the contrary, a MAM resident be a disease associated to alterations of MAM has started to emerge
fraction of the tumor suppressor p53 physically interacts with, as an interesting possibility. The discovery that both PS1 and PS2
and enhances the activity of, the Ca2+ -ATPase SERCA, increasing are enriched in MAM [27], as well as APP, was the first hit in sup-
ER [Ca2+ ] and sensitizing cells to apoptosis [153]. In addition to port of this idea. The presence of PSs, APP and ␥-secretase activity
the above mentioned effects on IP3Rs, Bcl-2 has also been reported in MAM has been independently confirmed by others [161], and
to interact with SERCA and limit its activity [154]. The oncogene we recently reported that FAD-PS2 mutants are enriched in MAM,
AKT (also known as PKB) has been reported to phosphorylate compared to the wt protein [30].
IP3Rs [155], inhibiting ER Ca2+ release and negatively affecting More debated is the role of PSs in ER-mitochondria con-
the Ca2+ -dependent apoptotic response [123,156]. On the contrary, tact sites. We firstly reported that the expression of the only
the tumor suppressor protein phosphatase and tensin homolog PS2 (wt and, more potently, FAD-mutants), but not of PS1,
(PTEN), commonly lost or mutated in human cancers, is one of the favors ER-mitochondria physical and functional coupling, result-
most important negative regulator of the AKT pathway. Indeed, not ing in an increased ER-mitochondria Ca2+ transfer [28]. Later,
only it dephosphorylates phosphatidylinositol 3,4,5-trisphosphate both endogenous PS1 and PS2 have been suggested to negatively
(PIP3) to phosphatidylinositol 4,5-bisphosphate (PIP2), via its lipid modulate ER-mitochondria coupling, while their FAD-mutants
phosphatase activity (thus blocking PIP3-dependent AKT recruit- increase organelles’ apposition [162], affecting phospholipids and
ment/activation to plasma membrane), but also directly interacts cholesterol metabolism. In this latter paper, an increased ER-
with IP3Rs and removes their AKT-dependent phosphorylation mitochondria coupling has been observed in fibroblasts from both
[125]. This latter effect is exerted specifically by an ER-resident FAD and sporadic AD patients. Though the observation is poten-
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 11

tially interesting, in our opinion it is not clear how sporadic AD platform. A long term A␤ exposure appears as a possible path-
cases may converge on MAM alterations similarly to those mecha- way, though further investigations will be necessary to address this
nistically caused by PSs or APP mutations, as it has been proposed crucial point.
[162]. Moreover, we have recently confirmed that only PS2 is able to
increase ER-mitochondria tethering, by binding and sequestering 4.4. Parkinson’s disease
Mfn2 [30], thus removing the inhibitory effect of the latter protein
on organelles’ apposition [8]. In addition, in another study, neu- Among neurodegenerative diseases, Parkinson’s Disease (PD) is
ronal FAD-PS1 expression has been associated with a decreased probably the first one for which genetic studies suggested, since the
ER-mitochondria physical and functional coupling [163]. beginning, a strong and quite immediate correlation with mito-
Several other evidence supports the link between ER- chondrial alterations. Indeed, some of the proteins mutated in
mitochondria association and AD. For example, acute exposure to familial forms of PD, such as PINK-1 and Parkin, are targeted to
pathogenic levels of A␤ has been reported to strengthen ER to mito- or dynamically associated with mitochondria [173]. More recent
chondria Ca2+ transfer [164] and an increased ER-mitochondria observations suggest that some of the PD-related proteins (not
coupling has been observed in different AD models [29,164]. Very only those classically associated with mitochondrial functionality,
recently, it has been shown that Mfn2 down-regulation, likely by see below) may be involved in the modulation of ER-mitochondria
increasing ER-mitochondria association, as verified by quantita- apposition, thus suggesting the intriguing possibility that, similarly
tive EM, results in a substantial alteration in the process of A␤ to AD, even some of the alterations observed in PD could be linked
generation [44]. This defect was associated to an impairment of to MAM anomalies. In many cases, however, though the functions
␥-secretase maturation. of wt endogenous proteins have been elucidated, studies on the
The relationship between A␤ peptides and mitochondria effects of PD-related mutants are still lacking.
remains also unclear. A␤ accumulation into mitochondria has been Endogenous Parkin, for instance, has been reported to favor
reported and suggested to increase ROS production and impair ER-mitochondria physical and functional coupling, increasing
mitochondrial respiration [165]. A multitude of mitochondrial mitochondrial Ca2+ uptake and sustaining ATP synthesis [174].
functions have been reported to be affected by A␤ [166,167]. Pro- Recently, however, a strengthened ER-mitochondria apposition has
vided that the machinery for A␤ production is not present in been described in fibroblasts from Parkin (PARK-2) knock-out mice
mitochondria, A␤ has been proposed to be taken up by mitochon- and from PD patients harboring PARK-2 mutations [175]. Higher
dria via a TIM/TOM-mediated (but -independent) mechanism levels of Mfn2 (a target protein ubiquitinated by Parkin) were
and observed, by immunoelectron microscopy, to localize to mito- observed and interpreted as responsible for the increased inter-
chondrial cristae [168]. A specific effect of A␤ on the processing organelle association. In our opinion, being the list of Parkin’s
of most N-terminal presequences of mitochondrial proteins has targets quite wide, the effects of Parkin ablation on the tether-
been reported and suggested to induce an imbalanced organel- ing may, or may not, be Mfn2-dependent. Similarly, drosophila
lar proteome, resulting in mitochondrial dysfunctions [169]. Very PINK1/Parkin mutants displayed defective mitochondria that acti-
recently, an elegant study failed to find an A␤ import activity into vate the PERK branch of UPR, a neurotoxic process potentially
mitochondria, but confirmed A␤ association (especially of A␤42) modulated by Mfn2 [176]. ␣-synuclein has been also reported to
with the organelle, likely due to OMM interactions [170]. Though favor mitochondrial Ca2+ uptake, by enhancing ER-mitochondria
the study was performed on isolated mitochondria, potentially los- physical association [177]. Recently, ␣-synuclein has been reported
ing some additional molecular components that may impact on the to be localized at MAM [178]: the disease-linked mutants are less
in vivo outcome, A␤ was not found to directly affect mitochondrial associated with MAM and induce ER-mitochondria uncoupling.
functionalities, but to impair mitochondrial pre-proteins import Overexpression of wt DJ-1 (a protein usually localized in the cyto-
within the organelle, because of its aggregation with these pro- sol − but see also below − whose mutant forms are associated with
teins [170]. However, beyond the specific effects on mitochondria, familial PD) has been also shown to increase ER-mitochondria cou-
it is still unclear how A␤ reaches these organelles. Indeed, after its pling [179]. The final outcome is that DJ-1 is able to counteract the
generation from plasma membrane-located APP, A␤ is released in negative effects of the tumor suppressor protein p53 on organelles
the extracellular environment or, in case of intracellular genera- juxtaposition [179]. Recently, it has been shown that, upon star-
tion, as it has been suggested [171], into the lumen of the involved vation, a fraction of DJ-1 translocates into mitochondrial matrix to
organelles. Even in the case of A␤ endocytosis from the extracel- sustain ATP synthesis, an ability which is lost by pathogenic DJ-1
lular milieu [172], how peptides can reach the cytosol before to mutants [180].
interact with mitochondria remains elusive. The fact that A␤ can
be produced in MAM (i.e., in close proximity to mitochondria) may 4.5. Amyotrophic lateral sclerosis
help to elucidate this aspect, but the topological problem discussed
above remains, to date, an open question. Overall, we believe that, Amyotrophic Lateral Sclerosis (ALS) is the most common form
though it is difficult to reconcile all these recent findings in one of motor neuron disease and is clinically and genetically associated
mechanistic model, it starts to be clear that AD and MAM alter- to Fronto-Temporal Dementia (FTD) [181]. Mutations in the genes
ations are associated. In particular, reports from different groups encoding for TDP-43, FUS and C9ORF72-derived protein are associ-
create consensus on the fact that ER-mitochondria coupling is early ated to ∼ 50% of the familial forms of the disease [181]. In neurons
increased in AD pathogenesis. Whether this increased association and glia of patients, cytosolic and nuclear inclusions of these pro-
has a direct impact on AD onset/progression, or it is just an epiphe- teins are observed. A key role for ER-mitochondria axis in ALS/FTD
nomenon, or an attempt to protect cells from other insults, is still onset/progression has been recently started to emerge (reviewed
unclear. In both cases, however, MAM modulation appears as a in [182]). In particular, overexpression of both wt and mutated
promising novel therapeutic target to exploit for pharmacologi- TDP-43 decreases ER-mitochondria physical and functional cou-
cal purpose. To this aim, the clarification of the precise molecular pling [20], by disrupting VAPB-PTPIP51 interaction (see above)
mechanism, that leads to an increased ER-mitochondria coupling, through activation of GSK-3␤. With a very similar molecular mech-
appears of fundamental importance. For instance, while FAD-PS2 anism, converging on a GSK-3␤-dependent disassembling of the
mutants expression seems able to directly affect the tethering, it VAPB-PTPIP51 tethering complex, FUS (wt and ALS-linked mutants)
is still not clear how other FAD-mutants, or still unknown molecu- overexpression has been reported to impair ER-mitochondria jux-
lar mechanisms in sporadic AD, may converge on ER-mitochondria taposition, Ca2+ transfer and, consequently, mitochondrial ATP
12 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

production [21]. Importantly, expression of VAPB mutant forms [12] S.E. Rice, V.I. Gelfand, Paradigm lost: milton connects kinesin heavy chain to
(associated with some familial forms of ALS type-8) increases miro on mitochondria, J. Cell Biol. 173 (2006) 459–461.
[13] S. Lahiri, J.T. Chao, S. Tavassoli, A.K. Wong, V. Choudhary, B.P. Young, C.J.
ER-mitochondria Ca2+ shuttling [19]. In mouse embryonic motor Loewen, W.A. Prinz, A conserved endoplasmic reticulum membrane protein
neurons, overexpression of the ALS-related hSOD1 mutant G93A, complex (EMC) facilitates phospholipid transfer from the ER to
has been observed to alter ER and mitochondrial Ca2+ homeostasis mitochondria, PLoS Biol. 12 (2014) e1001969.
[14] T. Satoh, A. Ohba, Z. Liu, T. Inagaki, A.K. Satoh, dPob/EMC is essential for
[183]. Moreover, loss of the MAM protein SIGMA1-R (a condition biosynthesis of rhodopsin and other multi-pass membrane proteins in
linked to some familial forms of ALS/FTD) has been reported to Drosophila photoreceptors, Elife 4 (2015).
impair ER-mitochondria communication [184], similarly to Dis- [15] J.G. Wideman, The ubiquitous and ancient ER membrane protein complex
(EMC): tether or not? F1000Res 4 (2015) 624.
tal Hereditary Motor Neuropathies (dHMNs)-associated SIGMA1-R
[16] A. Murley, R.D. Sarsam, A. Toulmay, J. Yamada, W.A. Prinz, J. Nunnari, Ltc1 is
loss-of-function mutations [185]. Overall, these studies strongly an ER-localized sterol transporter and a component of ER-mitochondria and
suggest that alterations of ER-mitochondria platform could be an ER-vacuole contacts, J. Cell Biol. 209 (2015) 539–548.
[17] T. Hayashi, R. Rizzuto, G. Hajnoczky, T.P. Su, MAM: more than just a
early event at the basis of ALS/FTD.
housekeeper, Trends Cell Biol. 19 (2009) 81–88.
[18] G. Szabadkai, K. Bianchi, P. Varnai, D. De Stefani, M.R. Wieckowski, D.
Cavagna, A.I. Nagy, T. Balla, R. Rizzuto, Chaperone-mediated coupling of
5. Conclusion endoplasmic reticulum and mitochondrial Ca2+ channels, J. Cell Biol. 175
(2006) 901–911.
ER-mitochondria coupling is increasingly suggested to play [19] K.J. De Vos, G.M. Morotz, R. Stoica, E.L. Tudor, K.F. Lau, S. Ackerley, A. Warley,
C.E. Shaw, C.C. Miller, VAPB interacts with the mitochondrial protein
essential roles in different fundamental cellular pathways. The
PTPIP51 to regulate calcium homeostasis, Hum. Mol. Genet. 21 (2012)
functional significances of this apposition have started to be 1299–1311.
elucidated and are constantly growing. Given that alterations of ER- [20] R. Stoica, K.J. De Vos, S. Paillusson, S. Mueller, R.M. Sancho, K.F. Lau, G.
mitochondria juxtaposition and/or cross-talk have been reported Vizcay-Barrena, W.L. Lin, Y.F. Xu, J. Lewis, D.W. Dickson, L. Petrucelli, J.C.
Mitchell, C.E. Shaw, C.C. Miller, ER-mitochondria associations are regulated
to be early observed in different pathological conditions, the mod- by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated
ulation of this key intracellular axis appears as a good candidate TDP-43, Nat. Commun. 5 (2014) 3996.
for possible therapeutic strategies, aimed at hindering the onset [21] R. Stoica, S. Paillusson, P. Gomez-Suaga, J.C. Mitchell, D.H. Lau, E.H. Gray,
R.M. Sancho, G. Vizcay-Barrena, K.J. De Vos, C.E. Shaw, D.P. Hanger, W.
and progression of devastating diseases. For this purpose, the exact Noble, C.C. Miller, ALS/FTD-associated FUS activates GSK-3beta to disrupt
knowledge of the molecules that control inter-organelle apposi- the VAPB-PTPIP51 interaction and ER-mitochondria associations, EMBO
tion appears of fundamental importance, and basic-science studies Rep. (2016).
[22] R. Galmes, A. Houcine, A.R. van Vliet, P. Agostinis, C.L. Jackson, F. Giordano,
seeking at defining their identity are encouraged. ORP5/ORP8 localize to endoplasmic reticulum-mitochondria contacts and
are involved in mitochondrial function, EMBO Rep. 17 (2016) 800–810.
[23] J. Chung, F. Torta, K. Masai, L. Lucast, H. Czapla, L.B. Tanner, P.
Acknowledgements Narayanas-wamy, M.R. Wenk, F. Nakatsu, P. De Camilli, INTRACELLULAR
TRANSPORT PI4P/phosphatidylserine countertransport at ORP5- and
The Authors thank grants from the University of Padova; the ORP8-mediated ER-plasma membrane contacts, Science 349 (2015)
428–432.
Italian Ministry of University and Scientific Research; Fondazione [24] T. Simmen, J.E. Aslan, A.D. Blagoveshchenskaya, L. Thomas, L. Wan, Y. Xiang,
Cassa di Risparmio di Padova e Rovigo (CARIPARO Foundation; S.F. Feliciangeli, C.H. Hung, C.M. Crump, G. Thomas, PACS-2 controls
Progetti d’eccellenza 2011/2012) and EU Joint Programme in endoplasmic reticulum-mitochondria communication and Bid-mediated
apoptosis, EMBO J. 24 (2005) 717–729.
Neurodegenerative Disease, 2015-2018, “Cellular Bioenergetics in [25] R.T. Youker, U. Shinde, R. Day, G. Thomas, At the crossroads of homoeostasis
Neurodegenerative Diseases: A system-based pathway and target and disease: roles of the PACS proteins in membrane traffic and apoptosis,
analysis” for their research work support. R.F. and P.T. are Research Biochem. J. 421 (2009) 1–15.
[26] R. Iwasawa, A.L. Mahul-Mellier, C. Datler, E. Pazarentzos, S. Grimm, Fis1 and
fellows of the CARIPARO and EU Joint Programme in Neurodegen-
Bap31 bridge the mitochondria-ER interface to establish a platform for
erative Disease grant, respectively. apoptosis induction, EMBO J. 30 (2011) 556–568.
[27] E. Area-Gomez, A.J. de Groof, I. Boldogh, T.D. Bird, G.E. Gibson, C.M. Koehler,
W.H. Yu, K.E. Duff, M.P. Yaffe, L.A. Pon, E.A. Schon, Presenilins are enriched in
References endoplasmic reticulum membranes associated with mitochondria, Am. J.
Pathol. 175 (2009) 1810–1816.
[1] W. Bernhard, C. Rouiller, Close topographical relationship between [28] E. Zampese, C. Fasolato, M.J. Kipanyula, M. Bortolozzi, T. Pozzan, P. Pizzo,
mitochondria and ergastoplasm of liver cells in a definite phase of cellular Presenilin 2 modulates endoplasmic reticulum (ER)-mitochondria
activity, J. Biophys. Biochem. Cytol. 2 (1956) 73–78. interactions and Ca2+ cross-talk, Proc. Natl. Acad. Sci. U. S. A. 108 (2011)
[2] R. Rizzuto, P. Pinton, W. Carrington, F.S. Fay, K.E. Fogarty, L.M. Lifshitz, R.A. 2777–2782.
Tuft, T. Pozzan, Close contacts with the endoplasmic reticulum as [29] M.J. Kipanyula, L. Contreras, E. Zampese, C. Lazzari, A.K. Wong, P. Pizzo, C.
determinants of mitochondrial Ca2+ responses, Science 280 (1998) Fasolato, T. Pozzan, Ca2+ dysregulation in neurons from transgenic mice
1763–1766. expressing mutant presenilin 2, Aging Cell 11 (2012) 885–893.
[3] C.A. Mannella, The relevance of mitochondrial membrane topology to [30] R. Filadi, E. Greotti, G. Turacchio, A. Luini, T. Pozzan, P. Pizzo, Presenilin 2
mitochondrial function, Biochim. Biophys. Acta 1762 (2006) 140–147. modulates endoplasmic reticulum-mitochondria coupling by tuning the
[4] A.A. Rowland, G.K. Voeltz, Endoplasmic reticulum-mitochondria contacts: antagonistic effect of mitofusin 2, Cell Rep. 15 (2016) 2226–2238.
function of the junction, Nat. Rev. Mol. Cell Biol. 13 (2012) 607–625. [31] O.M. de Brito, L. Scorrano, Mitofusin 2 tethers endoplasmic reticulum to
[5] S.J. Mueller, R. Reski, Mitochondrial dynamics and the ER, Front Cell Dev Biol mitochondria, Nature 456 (2008) 605–610.
3 (2015) 78. [32] T. Koshiba, S.A. Detmer, J.T. Kaiser, H. Chen, J.M. McCaffery, D.C. Chan,
[6] M. Giacomello, L. Pellegrini, The coming of age of the mitochondria-ER Structural basis of mitochondrial tethering by mitofusin complexes, Science
contact: a matter of thickness, Cell Death Differ. 23 (2016) 1417–1427. 305 (2004) 858–862.
[7] G. Csordas, C. Renken, P. Varnai, L. Walter, D. Weaver, K.F. Buttle, T. Balla, [33] N. Ishihara, Y. Eura, K. Mihara, Mitofusin 1 and 2 play distinct roles in
C.A. Mannella, G. Hajnoczky, Structural and functional features and mitochondrial fusion reactions via GTPase activity, J. Cell Sci. 117 (2004)
significance of the physical linkage between ER and mitochondria, J. Cell 6535–6546.
Biol. 174 (2006) 915–921. [34] M. Hamasaki, N. Furuta, A. Matsuda, A. Nezu, A. Yamamoto, N. Fujita, H.
[8] R. Filadi, E. Greotti, G. Turacchio, A. Luini, T. Pozzan, P. Pizzo, Mitofusin 2 Oomori, T. Noda, T. Haraguchi, Y. Hiraoka, A. Amano, T. Yoshimori,
ablation increases endoplasmic reticulum-mitochondria coupling, Proc. Autophagosomes form at ER-mitochondria contact sites, Nature 495 (2013)
Natl. Acad. Sci. U. S. A. 112 (2015) E2174–2181. 389–393.
[9] B. Kornmann, E. Currie, S.R. Collins, M. Schuldiner, J. Nunnari, J.S. Weissman, [35] A. Sugiura, S. Nagashima, T. Tokuyama, T. Amo, Y. Matsuki, S. Ishido, Y. Kudo,
P. Walter, An ER-mitochondria tethering complex revealed by a synthetic H.M. McBride, T. Fukuda, N. Matsushita, R. Inatome, S. Yanagi, MITOL
biology screen, Science 325 (2009) 477–481. regulates endoplasmic reticulum-mitochondria contacts via Mitofusin2,
[10] B. Kornmann, C. Osman, P. Walter, The conserved GTPase Gem1 regulates Mol. Cell 51 (2013) 20–34.
endoplasmic reticulum-mitochondria connections, Proc. Natl. Acad. Sci. U. S. [36] D.W. Hailey, A.S. Rambold, P. Satpute-Krishnan, K. Mitra, R. Sougrat, P.K.
A. 108 (2011) 14151–14156. Kim, J. Lippincott-Schwartz, Mitochondria supply membranes for
[11] A. Lang, A.T. John Peter, B. Kornmann, ER-mitochondria contact sites in autophagosome biogenesis during starvation, Cell 141 (2010) 656–667.
yeast: beyond the myths of ERMES, Curr. Opin. Cell Biol. 35 (2015) 7–12.
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 13

[37] A.S. Rambold, S. Cohen, J. Lippincott-Schwartz, Fatty acid trafficking in mediate tether assembly, Proc. Natl. Acad. Sci. U. S. A. 112 (2015)
starved cells: regulation by lipid droplet lipolysis, autophagy, and E3179–3188.
mitochondrial fusion dynamics, Dev. Cell 32 (2015) 678–692. [64] G. Drin, J.M. von Filseck, A. Copic, New molecular mechanisms of
[38] Y. Chen, G. Csordas, C. Jowdy, T.G. Schneider, N. Csordas, W. Wang, Y. Liu, M. inter-organelle lipid transport, Biochem. Soc. Trans. 44 (2016) 486–492.
Kohlhaas, M. Meiser, S. Bergem, J.M. Nerbonne, G.W. Dorn, 2nd, C. Maack [65] Y. Elbaz-Alon, M. Eisenberg-Bord, V. Shinder, S.B. Stiller, E. Shimoni, N.
Mitofusin 2-containing mitochondrial-reticular microdomains direct rapid Wiedemann, T. Geiger, M. Schuldiner, Lam6 regulates the extent of contacts
cardiomyocyte bioenergetic responses via interorganelle Ca(2 +) crosstalk, between organelles, Cell Rep. 12 (2015) 7–14.
Circ. Res. 111 (2012) 863–875. [66] A.T. Gatta, L.H. Wong, Y.Y. Sere, D.M. Calderon-Norena, S. Cockcroft, A.K.
[39] R. Filadi, T. Pozzan, Generation and functions of second messengers Menon, T.P. Levine, A new family of StART domain proteins at membrane
microdomains, Cell Calcium 58 (2015) 405–414. contact sites has a role in ER-PM sterol transport, Elife 4 (2015).
[40] M. Schneeberger, M.O. Dietrich, D. Sebastian, M. Imbernon, C. Castano, A. [67] V.V. Flis, G. Daum, Lipid transport between the endoplasmic reticulum and
Garcia, Y. Esteban, A. Gonzalez-Franquesa, I.C. Rodriguez, A. Bortolozzi, P.M. mitochondria, Cold Spring Harb. Perspect. Biol. 5 (2013).
Garcia-Roves, R. Gomis, R. Nogueiras, T.L. Horvath, A. Zorzano, M. Claret, [68] T.R. Guilarte, M.K. Loth, S.R. Guariglia, TSPO finds NOX2 in microglia for
Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and redox homeostasis, Trends Pharmacol. Sci. (2016).
energy imbalance, Cell 155 (2013) 172–187. [69] J.R. Friedman, L.L. Lackner, M. West, J.R. DiBenedetto, J. Nunnari, G.K. Voeltz,
[41] P. Cosson, A. Marchetti, M. Ravazzola, L. Orci, Mitofusin-2 independent ER tubules mark sites of mitochondrial division, Science 334 (2011)
juxtaposition of endoplasmic reticulum and mitochondria: an 358–362.
ultrastructural study, PLoS One 7 (2012) e46293. [70] T. Klecker, S. Bockler, B. Westermann, Making connections: interorganelle
[42] P.T. Wang, P.O. Garcin, M. Fu, M. Masoudi, P. St-Pierre, N. Pante, I.R. Nabi, contacts orchestrate mitochondrial behavior, Trends Cell Biol. 24 (2014)
Distinct mechanisms controlling rough and smooth endoplasmic reticulum 537–545.
contacts with mitochondria, J. Cell Sci. 128 (2015) 2759–2765. [71] F. Korobova, V. Ramabhadran, H.N. Higgs, An actin-dependent step in
[43] L. Li, G. Gao, J. Shankar, B. Joshi, L.J. Foster, I.R. Nabi, p38 MAP mitochondrial fission mediated by the ER-associated formin INF2, Science
kinase-dependent phosphorylation of the Gp78 E3 ubiquitin ligase controls (New York, N.Y.) 339 (2013) 464–467.
ER-mitochondria association and mitochondria motility, Mol. Biol. Cell. 26 [72] J. Prudent, H.M. McBride, Mitochondrial dynamics ER actin tightens the drp1
(2015) 3828–3840. noose, Curr. Biol. 26 (2016) R207–209.
[44] N.S. Leal, B. Schreiner, C.M. Pinho, R. Filadi, B. Wiehager, H. Karlstrom, P. [73] B. Westermann, Mitochondrial fusion and fission in cell life and death, Nat.
Pizzo, M. Ankarcrona, Mitofusin-2 knockdown increases ER-mitochondria Rev. Mol. Cell Biol. 11 (2010) 872–884.
contact and decreases amyloid beta-peptide production, J. Cell. Mol. Med. [74] T. Garofalo, P. Matarrese, V. Manganelli, M. Marconi, A. Tinari, L.
(2016). Gambardella, A. Faggioni, R. Misasi, M. Sorice, W. Malorni, Evidence for the
[45] D. Naon, M. Zaninello, M. Giacomello, T. Varanita, F. Grespi, S. involvement of lipid rafts localized at the ER-mitochondria associated
Lakshminaranayan, A. Serafini, M. Semenzato, S. Herkenne, M.I. membranes in autophagosome formation, Autophagy 12 (2016) 917–935.
Hernandez-Alvarez, A. Zorzano, D. De Stefani, G.W. Dorn 2nd, L. Scorrano, [75] S. Luo, Q. Chen, E. Cebollero, D. Xing, Mitochondria: one of the origins for
Critical reappraisal confirms that Mitofusin 2 is an endoplasmic autophagosomal membranes? Mitochondrion 9 (2009) 227–231.
reticulum-mitochondria tether, Proc. Natl. Acad. Sci. U. S. A. 113 (2016) [76] L. Liu, D. Feng, G. Chen, M. Chen, Q. Zheng, P. Song, Q. Ma, C. Zhu, R. Wang,
11249–11254. W. Qi, L. Huang, P. Xue, B. Li, X. Wang, H. Jin, J. Wang, F. Yang, P. Liu, Y. Zhu, S.
[46] G. Csordas, P. Varnai, T. Golenar, S. Roy, G. Purkins, T.G. Schneider, T. Balla, G. Sui, Q. Chen, Mitochondrial outer-membrane protein FUNDC1 mediates
Hajnoczky, Imaging interorganelle contacts and local calcium dynamics at hypoxia-induced mitophagy in mammalian cells, Nat. Cell Biol. 14 (2012)
the ER-mitochondrial interface, Mol. Cell 39 (2010) 121–132. 177–185.
[47] W.M. Roberts, Localization of calcium signals by a mobile calcium buffer in [77] M. Chen, Z. Chen, Y. Wang, Z. Tan, C. Zhu, Y. Li, Z. Han, L. Chen, R. Gao, L. Liu,
frog saccular hair cells, J. Neurosci. 14 (1994) 3246–3262. Q. Chen, Mitophagy receptor FUNDC1 regulates mitochondrial dynamics
[48] H. Chen, S.A. Detmer, A.J. Ewald, E.E. Griffin, S.E. Fraser, D.C. Chan, Mitofusins and mitophagy, Autophagy 12 (2016) 689–702.
Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential [78] W. Wu, C. Lin, K. Wu, L. Jiang, X. Wang, W. Li, H. Zhuang, X. Zhang, H. Chen, S.
for embryonic development, J. Cell Biol. 160 (2003) 189–200. Li, Y. Yang, Y. Lu, J. Wang, R. Zhu, L. Zhang, S. Sui, N. Tan, B. Zhao, J. Zhang, L.
[49] A. Mourier, E. Motori, T. Brandt, M. Lagouge, I. Atanassov, A. Galinier, G. Li, D. Feng, FUNDC1 regulates mitochondrial dynamics at the
Rappl, S. Brodesser, K. Hultenby, C. Dieterich, N.G. Larsson, Mitofusin 2 is ER-mitochondrial contact site under hypoxic conditions, EMBO J. 35 (2016)
required to maintain mitochondrial coenzyme Q levels, J. Cell Biol. 208 1368–1384.
(2015) 429–442. [79] K. Schroder, J. Tschopp, The inflammasomes, Cell 140 (2010) 821–832.
[50] D. De Stefani, M. Patron, R. Rizzuto, Structure and function of the [80] R. Zhou, A.S. Yazdi, P. Menu, J. Tschopp, A role for mitochondria in NLRP3
mitochondrial calcium uniporter complex, Biochim. Biophys. Acta 1853 inflammasome activation, Nature 469 (2011) 221–225.
(2015) 2006–2011. [81] D.M. Booth, B. Enyedi, M. Geiszt, P. Varnai, G. Hajnoczky, Redox
[51] J.E. Vance, Phospholipid synthesis in a membrane fraction associated with nanodomains are induced by and control calcium signaling at the
mitochondria, J. Biol. Chem. 265 (1990) 7248–7256. ER-Mitochondrial interface, Mol. Cell 63 (2016) 240–248.
[52] A.E. Rusinol, Z. Cui, M.H. Chen, J.E. Vance, A unique mitochondria-associated [82] S. Marchi, S. Patergnani, P. Pinton, The endoplasmic reticulum-mitochondria
membrane fraction from rat liver has a high capacity for lipid synthesis and connection: one touch multiple functions, Biochim. Biophys. Acta 1837
contains pre-Golgi secretory proteins including nascent lipoproteins, J. Biol. (2014) 461–469.
Chem. 269 (1994) 27494–27502. [83] A.R. van Vliet, T. Verfaillie, P. Agostinis, New functions of mitochondria
[53] S.J. Stone, J.E. Vance, Phosphatidylserine synthase-1 and −2 are localized to associated membranes in cellular signaling, Biochim. Biophys. Acta 1843
mitochondria-associated membranes, J. Biol. Chem. 275 (2000) (2014) 2253–2262.
34534–34540. [84] M. Schrader, L.F. Godinho, J.L. Costello, M. Islinger, The different facets of
[54] J. Zborowski, A. Dygas, L. Wojtczak, Phosphatidylserine decarboxylase is organelle interplay-an overview of organelle interactions, Front. Cell Dev.
located on the external side of the inner mitochondrial membrane, FEBS Biol. 3 (2015) 56.
Lett. 157 (1983) 179–182. [85] A. Rimessi, V. Bezzerri, S. Patergnani, S. Marchi, G. Cabrini, P. Pinton,
[55] C. Osman, D.R. Voelker, T. Langer, Making heads or tails of phospholipids in Mitochondrial Ca2+-dependent NLRP3 activation exacerbates the
mitochondria, J. Cell Biol. 192 (2011) 7–16. Pseudomonas aeruginosa-driven inflammatory response in cystic fibrosis,
[56] D.R. Voelker, Interorganelle transport of aminoglycerophospholipids, Nat. Commun. 6 (2015) 6201.
Biochim. Biophys. Acta 1486 (2000) 97–107. [86] A. Murley, L.L. Lackner, C. Osman, M. West, G.K. Voeltz, P. Walter, J. Nunnari,
[57] J.E. Vance, MAM(mitochondria-associated membranes) in mammalian cells: ER-associated mitochondrial division links the distribution of mitochondria
lipids and beyond, Biochim. Biophys. Acta 1841 (2014) 595–609. and mitochondrial DNA in yeast, Elife 2 (2013) e00422.
[58] J. Stiban, L. Caputo, M. Colombini, Ceramide synthesis in the endoplasmic [87] S.C. Lewis, L.F. Uchiyama, J. Nunnari, ER-mitochondria contacts couple
reticulum can permeabilize mitochondria to proapoptotic proteins, J. Lipid mtDNA synthesis with mitochondrial division in human cells, Science 353
Res. 49 (2008) 625–634. (2016) aaf5549.
[59] T. Hayashi, M. Fujimoto, Detergent-resistant microdomains determine the [88] H.F. Deluca, G.W. Engstrom, Calcium uptake by rat kidney mitochondria,
localization of sigma-1 receptors to the endoplasmic Proc. Natl. Acad. Sci. U. S. A. 47 (1961) 1744–1750.
reticulum-mitochondria junction, Mol. Pharmacol. 77 (2010) 517–528. [89] P. Mitchell, J. Moyle, Chemiosmotic hypothesis of oxidative
[60] A. Murley, J. Nunnari, The emerging network of mitochondria-organelle phosphorylation, Nature 213 (1967) 137–139.
contacts, Mol. Cell 61 (2016) 648–653. [90] M. Bragadin, T. Pozzan, G.F. Azzone, Activation energies and enthalpies
[61] K.O. Kopec, V. Alva, A.N. Lupas, Homology of SMP domains to the TULIP during Ca2+ transport in rat liver mitochondria, FEBS Lett. 104 (1979)
superfamily of lipid-binding proteins provides a structural basis for lipid 347–351.
exchange between ER and mitochondria, Bioinformatics 26 (2010) [91] J.M. Baughman, F. Perocchi, H.S. Girgis, M. Plovanich, C.A. Belcher-Timme, Y.
1927–1931. Sancak, X.R. Bao, L. Strittmatter, O. Goldberger, R.L. Bogorad, V. Koteliansky,
[62] K. Maeda, K. Anand, A. Chiapparino, A. Kumar, M. Poletto, M. Kaksonen, A.C. V.K. Mootha, Integrative genomics identifies MCU as an essential component
Gavin, Interactome map uncovers phosphatidylserine transport by of the mitochondrial calcium uniporter, Nature 476 (2011) 341–345.
oxysterol-binding proteins, Nature 501 (2013) 257–261. [92] D. De Stefani, A. Raffaello, E. Teardo, I. Szabo, R. Rizzuto, A forty-kilodalton
[63] A.P. AhYoung, J. Jiang, J. Zhang, X. Khoi Dang, J.A. Loo, Z.H. Zhou, P.F. Egea, protein of the inner membrane is the mitochondrial calcium uniporter,
Conserved SMP domains of the ERMES complex bind phospholipids and Nature (2011).
14 R. Filadi et al. / Cell Calcium 62 (2017) 1–15

[93] D. De Stefani, R. Rizzuto, T. Pozzan, Enjoy the trip: calcium in mitochondria [120] P.J. Bartlett, L.D. Gaspers, N. Pierobon, A.P. Thomas, Calcium-dependent
back and forth, Annu. Rev. Biochem. 85 (2016) 161–192. regulation of glucose homeostasis in the liver, Cell Calcium 55 (2014)
[94] C. Mammucari, A. Raffaello, D. Vecellio Reane, R. Rizzuto, Molecular 306–316.
structure and pathophysiological roles of the Mitochondrial Calcium [121] L. Ozcan, C.C. Wong, G. Li, T. Xu, U. Pajvani, S.K. Park, A. Wronska, B.X. Chen,
Uniporter, Biochim. Biophys. Acta 1863 (2016) 2457–2464. A.R. Marks, A. Fukamizu, J. Backs, H.A. Singer, J.R. Yates 3rd, D. Accili, I. Tabas,
[95] K.J. Kamer, V.K. Mootha, The molecular era of the mitochondrial calcium Calcium signaling through CaMKII regulates hepatic glucose production in
uniporter, Nat. Rev. Mol. Cell Biol. 16 (2015) 545–553. fasting and obesity, Cell Metab. 15 (2012) 739–751.
[96] R. Rizzuto, A.W. Simpson, M. Brini, T. Pozzan, Rapid changes of [122] Y. Wang, G. Li, J. Goode, J.C. Paz, K. Ouyang, R. Screaton, W.H. Fischer, J. Chen,
mitochondrial Ca2+ revealed by specifically targeted recombinant aequorin, I. Tabas, M. Montminy, Inositol-1,4,5-trisphosphate receptor regulates
Nature 358 (1992) 325–327. hepatic gluconeogenesis in fasting and diabetes, Nature 485 (2012)
[97] D. Pendin, E. Greotti, R. Filadi, T. Pozzan, Spying on organelle Ca2+ in living 128–132.
cells: the mitochondrial point of view, J. Endocrinol. Invest. (2014). [123] T. Szado, V. Vanderheyden, J.B. Parys, H. De Smedt, K. Rietdorf, L. Kotelevets,
[98] G. Csordás, A.P. Thomas, G. Hajnoczky, Quasi-synaptic calcium signal E. Chastre, F. Khan, U. Landegren, O. Soderberg, M.D. Bootman, H.L. Roderick,
transmission between endoplasmic reticulum and mitochondria, EMBO J. 18 Phosphorylation of inositol 1,4,5-trisphosphate receptors by protein kinase
(1999) 96–108. B/Akt inhibits Ca2+ release and apoptosis, Proc, Natl. Acad. Sci. U. S. A. 105
[99] M. Giacomello, I. Drago, M. Bortolozzi, M. Scorzeto, A. Gianelle, P. Pizzo, T. (2008) 2427–2432.
Pozzan, Ca2+ hot spots on the mitochondrial surface are generated by Ca2+ [124] C. Betz, D. Stracka, C. Prescianotto-Baschong, M. Frieden, N. Demaurex, M.N.
mobilization from stores, but not by activation of store-operated Ca2+ Hall, Feature Article: mTOR complex 2-Akt signaling at
channels, Mol. Cell 38 (2010) 280–290. mitochondria-associated endoplasmic reticulum membranes (MAM)
[100] P. Pizzo, I. Drago, R. Filadi, T. Pozzan, Mitochondrial Ca2+ homeostasis: regulates mitochondrial physiology, Proc. Natl. Acad. Sci. U. S. A. 110 (2013)
mechanism, role, and tissue specificities, Pflugers Archiv: European Journal 12526–12534.
of Physiology 464 (2012) 3–17. [125] A. Bononi, M. Bonora, S. Marchi, S. Missiroli, F. Poletti, C. Giorgi, P.P. Pandolfi,
[101] L.S. Jouaville, P. Pinton, C. Bastianutto, G.A. Rutter, R. Rizzuto, Regulation of P. Pinton, Identification of PTEN at the ER and MAMs and its regulation of
mitochondrial ATP synthesis by calcium: evidence for a long-term Ca(2+) signaling and apoptosis in a protein phosphatase-dependent manner,
metabolic priming, Proc. Natl. Acad. Sci. U. S. A. 96 (1999) 13807–13812. Cell Death Differ. 20 (2013) 1631–1643.
[102] J.G. Pitter, P. Maechler, C.B. Wollheim, A. Spat, Mitochondria respond to [126] E. Tubbs, P. Theurey, G. Vial, N. Bendridi, A. Bravard, M.A. Chauvin, J. Ji-Cao,
Ca2+ already in the submicromolar range: correlation with redox state, Cell F. Zoulim, B. Bartosch, M. Ovize, H. Vidal, J. Rieusset,
Calcium 31 (2002) 97–104. Mitochondria-associated endoplasmic reticulum membrane (MAM)
[103] R.M. Denton, Regulation of mitochondrial dehydrogenases by calcium ions, integrity is required for insulin signaling and is implicated in hepatic insulin
Biochim. Biophys. Acta 1787 (2009) 1309–1316. resistance, Diabetes 63 (2014) 3279–3294.
[104] R.M. Denton, D.A. Richards, J.G. Chin, Calcium ions and the regulation of [127] T. Cali, D. Ottolini, M. Brini, Calcium and endoplasmic
NAD+-linked isocitrate dehydrogenase from the mitochondria of rat heart reticulum-mitochondria tethering in neurodegeneration, DNA Cell Biol. 32
and other tissues, Biochem. J. 176 (1978) 899–906. (2013) 140–146.
[105] P.R. Territo, V.K. Mootha, S.A. French, R.S. Balaban, Ca(2+) activation of heart [128] C. Lopez-Crisosto, R. Bravo-Sagua, M. Rodriguez-Pena, C. Mera, P.F. Castro,
mitochondrial oxidative phosphorylation: role of the F(0)/F(1)-ATPase, Am. A.F. Quest, B.A. Rothermel, M. Cifuentes, S. Lavandero, ER-to-mitochondria
J. Physiol. Cell Physiol. 278 (2000) C423–435. miscommunication and metabolic diseases, Biochim. Biophys. Acta 1852
[106] B. Glancy, W.T. Willis, D.J. Chess, R.S. Balaban, Effect of calcium on the (2015) 2096–2105.
oxidative phosphorylation cascade in skeletal muscle mitochondria, [129] S. Marchi, P. Pinton, Alterations of calcium homeostasis in cancer cells, Curr.
Biochemistry 52 (2013) 2793–2809. Opin. Pharmacol. 29 (2016) 1–6.
[107] A. del Arco, J. Satrustegui, Identification of a novel human subfamily of [130] U. Ozcan, Q. Cao, E. Yilmaz, A.H. Lee, N.N. Iwakoshi, E. Ozdelen, G. Tuncman,
mitochondrial carriers with calcium-binding domains, J. Biol. Chem. 279 C. Gorgun, L.H. Glimcher, G.S. Hotamisligil, Endoplasmic reticulum stress
(2004) 24701–24713. links obesity, insulin action, and type 2 diabetes, Science 306 (2004)
[108] J. Satrustegui, B. Pardo, A. Del Arco, Mitochondrial transporters as novel 457–461.
targets for intracellular calcium signaling, Physiol. Rev. 87 (2007) 29–67. [131] C. Bonnard, A. Durand, S. Peyrol, E. Chanseaume, M.A. Chauvin, B. Morio, H.
[109] C. Cardenas, R.A. Miller, I. Smith, T. Bui, J. Molgo, M. Muller, H. Vais, K.H. Vidal, J. Rieusset, Mitochondrial dysfunction results from oxidative stress in
Cheung, J. Yang, I. Parker, C.B. Thompson, M.J. Birnbaum, K.R. Hallows, J.K. the skeletal muscle of diet-induced insulin-resistant mice, J. Clin. Invest. 118
Foskett, Essential regulation of cell bioenergetics by constitutive InsP3 (2008) 789–800.
receptor Ca2+ transfer to mitochondria, Cell 142 (2010) 270–283. [132] J. Rieusset, Contribution of mitochondria and endoplasmic reticulum
[110] K. Mallilankaraman, C. Cardenas, P.J. Doonan, H.C. Chandramoorthy, K.M. dysfunction in insulin resistance: distinct or interrelated roles? Diab. Metab.
Irrinki, T. Golenar, G. Csordas, P. Madireddi, J. Yang, M. Muller, R. Miller, J.E. (2015).
Kolesar, J. Molgo, B. Kaufman, G. Hajnoczky, J.K. Foskett, M. Madesh, MCUR1 [133] A.P. Arruda, B.M. Pers, G. Parlakgul, E. Guney, K. Inouye, G.S. Hotamisligil,
is an essential component of mitochondrial Ca2+ uptake that regulates Chronic enrichment of hepatic endoplasmic reticulum-mitochondria
cellular metabolism, Nat. Cell Biol. 14 (2012) 1336–1343. contact leads to mitochondrial dysfunction in obesity, Nat. Med. 20 (2014)
[111] R. Bravo, J.M. Vicencio, V. Parra, R. Troncoso, J.P. Munoz, M. Bui, C. Quiroga, 1427–1435.
A.E. Rodriguez, H.E. Verdejo, J. Ferreira, M. Iglewski, M. Chiong, T. Simmen, [134] P. Theurey, J. Rieusset, Mitochondria-associated membranes response to
A. Zorzano, J.A. Hill, B.A. Rothermel, G. Szabadkai, S. Lavandero, Increased nutrient availability and role in metabolic diseases, Trends Endocrinol.
ER-mitochondrial coupling promotes mitochondrial respiration and Metab. (2016).
bioenergetics during early phases of ER stress, J. Cell Sci. 124 (2011) [135] A. Hagiwara, M. Cornu, N. Cybulski, P. Polak, C. Betz, F. Trapani, L.
2143–2152. Terracciano, M.H. Heim, M.A. Ruegg, M.N. Hall, Hepatic mTORC2 activates
[112] A. Rasola, P. Bernardi, Mitochondrial permeability transition in glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c, Cell
Ca2+-dependent apoptosis and necrosis, Cell Calcium (2011). Metab. 15 (2012) 725–738.
[113] C. Giorgi, F. Baldassari, A. Bononi, M. Bonora, E. De Marchi, S. Marchi, S. [136] D. Sebastian, M.I. Hernandez-Alvarez, J. Segales, E. Sorianello, J.P. Munoz, D.
Missiroli, S. Patergnani, A. Rimessi, J.M. Suski, M.R. Wieckowski, P. Pinton, Sala, A. Waget, M. Liesa, J.C. Paz, P. Gopalacharyulu, M. Oresic, S. Pich, R.
Mitochondrial Ca(2 +) and apoptosis, Cell Calcium 52 (2012) 36–43. Burcelin, M. Palacin, A. Zorzano, Mitofusin 2 (Mfn2) links mitochondrial and
[114] M. Bonora, A. Bononi, E. De Marchi, C. Giorgi, M. Lebiedzinska, S. Marchi, S. endoplasmic reticulum function with insulin signaling and is essential for
Patergnani, A. Rimessi, J.M. Suski, A. Wojtala, M.R. Wieckowski, G. Kroemer, normal glucose homeostasis, Proc. Natl. Acad. Sci. U. S. A. 109 (2012)
L. Galluzzi, P. Pinton, Role of the c subunit of the FO ATP synthase in 5523–5528.
mitochondrial permeability transition, ABBV Cell Cycle 12 (2013) 674–683. [137] L. Ozcan, J. Cristina de Souza, A.A. Harari, J. Backs, E.N. Olson, I. Tabas,
[115] V. Giorgio, S. von Stockum, M. Antoniel, A. Fabbro, F. Fogolari, M. Forte, G.D. Activation of calcium/calmodulin-dependent protein kinase II in obesity
Glick, V. Petronilli, M. Zoratti, I. Szabo, G. Lippe, P. Bernardi, Dimers of mediates suppression of hepatic insulin signaling, Cell Metab. 18 (2013)
mitochondrial ATP synthase form the permeability transition pore, Proc. 803–815.
Natl. Acad. Sci. U. S. A. 110 (2013) 5887–5892. [138] J.H. Lim, H.J. Lee, M. Ho Jung, J. Song, Coupling mitochondrial dysfunction to
[116] G. Morciano, C. Giorgi, M. Bonora, S. Punzetti, R. Pavasini, M.R. Wieckowski, endoplasmic reticulum stress response: a molecular mechanism leading to
G. Campo, P. Pinton, Molecular identity of the mitochondrial permeability hepatic insulin resistance, Cell. Signal. 21 (2009) 169–177.
transition pore and its role in ischemia-reperfusion injury, J. Mol. Cell. [139] S. Fu, L. Yang, P. Li, O. Hofmann, L. Dicker, W. Hide, X. Lin, S.M. Watkins, A.R.
Cardiol. 78 (2015) 142–153. Ivanov, G.S. Hotamisligil, Aberrant lipid metabolism disrupts calcium
[117] M.S. Hwang, C.T. Schwall, E. Pazarentzos, C. Datler, N.N. Alder, S. Grimm, homeostasis causing liver endoplasmic reticulum stress in obesity, Nature
Mitochondrial Ca(2+) influx targets cardiolipin to disintegrate respiratory 473 (2011) 528–531.
chain complex II for cell death induction, Cell Death Differ. 21 (2014) [140] T. Gutierrez, V. Parra, R. Troncoso, C. Pennanen, A. Contreras-Ferrat, C.
1733–1745. Vasquez-Trincado, P.E. Morales, C. Lopez-Crisosto, C. Sotomayor-Flores, M.
[118] A. Rimessi, S. Patergnani, M. Bonora, M.R. Wieckowski, P. Pinton, Chiong, B.A. Rothermel, S. Lavandero, Alteration in mitochondrial Ca(2+)
Mitochondrial Ca(2+) remodeling is a prime factor in oncogenic behavior, uptake disrupts insulin signaling in hypertrophic cardiomyocytes, Cell
Front. Oncol. 5 (2015) 143. Commun. Signal. 12 (2014) 68.
[119] M. Bittremieux, J.B. Parys, P. Pinton, G. Bultynck, ER functions of oncogenes [141] R. Ye, M. Ni, M. Wang, S. Luo, G. Zhu, R.H. Chow, A.S. Lee, Inositol
and tumor suppressors: modulators of intracellular Ca(2+) signaling, 1,4,5-trisphosphate receptor 1 mutation perturbs glucose homeostasis and
Biochim. Biophys. Acta 1863 (2016) 1364–1378.
R. Filadi et al. / Cell Calcium 62 (2017) 1–15 15

enhances susceptibility to diet-induced diabetes, J. Endocrinol. 210 (2011) [163] D. Sepulveda-Falla, A. Barrera-Ocampo, C. Hagel, A. Korwitz, M.F.
209–217. Vinueza-Veloz, K. Zhou, M. Schonewille, H. Zhou, L. Velazquez-Perez, R.
[142] M. Paillard, E. Tubbs, P.A. Thiebaut, L. Gomez, J. Fauconnier, C.C. Da Silva, G. Rodriguez-Labrada, A. Villegas, I. Ferrer, F. Lopera, T. Langer, C.I. De Zeeuw,
Teixeira, N. Mewton, E. Belaidi, A. Durand, M. Abrial, A. Lacampagne, J. M. Glatzel, Familial Alzheimer’s disease-associated presenilin-1 alters
Rieusset, M. Ovize, Depressing mitochondria-reticulum interactions cerebellar activity and calcium homeostasis, J. Clin. Invest. 124 (2014)
protects cardiomyocytes from lethal hypoxia-reoxygenation injury, 1552–1567.
Circulation 128 (2013) 1555–1565. [164] L. Hedskog, C.M. Pinho, R. Filadi, A. Ronnback, L. Hertwig, B. Wiehager, P.
[143] J. Rieusset, J. Fauconnier, M. Paillard, E. Belaidi, E. Tubbs, M.A. Chauvin, A. Larssen, S. Gellhaar, A. Sandebring, M. Westerlund, C. Graff, B. Winblad, D.
Durand, A. Bravard, G. Teixeira, B. Bartosch, M. Michelet, P. Theurey, G. Vial, Galter, H. Behbahani, P. Pizzo, E. Glaser, M. Ankarcrona, Modulation of the
M. Demion, E. Blond, F. Zoulim, L. Gomez, H. Vidal, A. Lacampagne, M. Ovize, endoplasmic reticulum-mitochondria interface in Alzheimer’s disease and
Disruption of calcium transfer from ER to mitochondria links alterations of related models, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 7916–7921.
mitochondria-associated ER membrane integrity to hepatic insulin [165] M. Manczak, T.S. Anekonda, E. Henson, B.S. Park, J. Quinn, P.H. Reddy,
resistance, Diabetologia 59 (2016) 614–623. Mitochondria are a direct site of Ab accumulation in Alzheimer’s disease
[144] C.H. Wang, Y.F. Chen, C.Y. Wu, P.C. Wu, Y.L. Huang, C.H. Kao, C.H. Lin, L.S. neurons: implications for free radical generation and oxidative damage in
Kao, T.F. Tsai, Y.H. Wei, Cisd2 modulates the differentiation and functioning disease progression, Hum. Mol. Genet. 15 (2006) 1437–1449.
of adipocytes by regulating intracellular Ca2+ homeostasis, Hum. Mol. [166] J.E. Selfridge, L.E.J. Lu, R.H. Swerdlow, Role of mitochondrial homeostasis
Genet. 23 (2014) 4770–4785. and dynamics in Alzheimer’s disease, Neurobiol. Dis. 51 (2013) 3–12.
[145] J.E. Galgani, L.K. Heilbronn, K. Azuma, D.E. Kelley, J.B. Albu, X. Pi-Sunyer, S.R. [167] V.A. Morais, B. De Strooper, Mitochondria dysfunction and
Smith, E. Ravussin, A.A.R.G. Look, Metabolic flexibility in response to glucose neurodegenerative disorders: cause or consequence, J. Alzheimers Dis. 20
is not impaired in people with type 2 diabetes after controlling for glucose (Suppl. 2) (2010) S255–263.
disposal rate, Diabetes 57 (2008) 841–845. [168] C.A. Hansson Petersen, N. Alikhani, H. Behbahani, B. Wiehager, P.F. Pavlov, I.
[146] D.M. Muoio, Metabolic inflexibility: when mitochondrial indecision leads to Alafuzoff, V. Leinonen, A. Ito, B. Winblad, E. Glaser, M. Ankarcrona, The
metabolic gridlock, Cell 159 (2014) 1253–1262. amyloid beta-peptide is imported into mitochondria via the TOM import
[147] A. Sood, D.V. Jeyaraju, J. Prudent, A. Caron, P. Lemieux, H.M. McBride, M. machinery and localized to mitochondrial cristae, Proc. Natl. Acad. Sci. U. S.
Laplante, K. Toth, L. Pellegrini, A Mitofusin-2-dependent inactivating A. 105 (2008) 13145–13150.
cleavage of Opa1 links changes in mitochondria cristae and ER contacts in [169] D. Mossmann, F.N. Vogtle, A.A. Taskin, P.F. Teixeira, J. Ring, J.M. Burkhart, N.
the postprandial liver, Proc. Natl. Acad. Sci. U. S. A. 111 (2014) 16017–16022. Burger, C.M. Pinho, J. Tadic, D. Loreth, C. Graff, F. Metzger, A. Sickmann, O.
[148] P. Theurey, E. Tubbs, G. Vial, J. Jacquemetton, N. Bendridi, M.A. Chauvin, M.R. Kretz, N. Wiedemann, R.P. Zahedi, F. Madeo, E. Glaser, C. Meisinger,
Alam, M. Le Romancer, H. Vidal, J. Rieusset, Mitochondria-associated Amyloid-beta peptide induces mitochondrial dysfunction by inhibition of
endoplasmic reticulum membranes allow adaptation of mitochondrial preprotein maturation, Cell Metab. 20 (2014) 662–669.
metabolism to glucose availability in the liver, J. Mol. Cell. Biol. (2016). [170] G. Cenini, C. Rub, M. Bruderek, W. Voos, Amyloid beta-peptides interfere
[149] T. Galbo, G.S. Olsen, B. Quistorff, E. Nishimura, Free fatty acid-induced PP2A with mitochondrial preprotein import competence by a coaggregation
hyperactivity selectively impairs hepatic insulin action on glucose process, Mol. Biol. Cell. 27 (2016) 3257–3272.
metabolism, PLoS One 6 (2011) e27424. [171] C. Haass, C. Kaether, G. Thinakaran, S. Sisodia, Trafficking and proteolytic
[150] F. Nardi, C. Lipina, D. Magill, R. Hage Hassan, E. Hajduch, A. Gray, H.S. processing of APP, Cold Spring Harb. Perspect. Med. 2 (2012) a006270.
Hundal, Enhanced insulin sensitivity associated with provision of mono and [172] L. Rajendran, W. Annaert, Membrane trafficking pathways in Alzheimer’s
polyunsaturated fatty acids in skeletal muscle cells involves counter disease, Traffic 13 (2012) 759–770.
modulation of PP2A, PLoS One 9 (2014) e92255. [173] T. Cali, D. Ottolini, M. Brini, Mitochondria, calcium, and endoplasmic
[151] T.A. Stewart, K.T. Yapa, G.R. Monteith, Altered calcium signaling in cancer reticulum stress in Parkinson’s disease, Biofactors 37 (2011) 228–240.
cells, Biochim. Biophys. Acta 1848 (2015) 2502–2511. [174] T. Cali, D. Ottolini, A. Negro, M. Brini, Enhanced parkin levels favor
[152] S. Marchi, C. Giorgi, M. Oparka, J. Duszynski, M.R. Wieckowski, P. Pinton, ER-mitochondria crosstalk and guarantee Ca(2+) transfer to sustain cell
Oncogenic and oncosuppressive signal transduction at bioenergetics, Biochim. Biophys. Acta 1832 (2013) 495–508.
mitochondria-associated endoplasmic reticulum membranes, Mol. Cell [175] C.A. Gautier, Z. Erpapazoglou, F. Mouton-Liger, M.P. Muriel, F. Cormier, S.
Oncol. 1 (2014) e956469. Bigou, S. Duffaure, M. Girard, B. Foret, A. Iannielli, V. Broccoli, C. Dalle, D.
[153] C. Giorgi, M. Bonora, G. Sorrentino, S. Missiroli, F. Poletti, J.M. Suski, F. Bohl, P.P. Michel, J.C. Corvol, A. Brice, O. Corti, The endoplasmic
Galindo Ramirez, R. Rizzuto, F. Di Virgilio, E. Zito, P.P. Pandolfi, M.R. reticulum-mitochondria interface is perturbed in PARK2 knockout mice and
Wieckowski, F. Mammano, G. Del Sal, P. Pinton, p53 at the endoplasmic patients with PARK2 mutations, Hum. Mol. Genet. (2016).
reticulum regulates apoptosis in a Ca2+-dependent manner, Proc. Natl. [176] I. Celardo, A.C. Costa, S. Lehmann, C. Jones, N. Wood, N.E. Mencacci, G.R.
Acad. Sci. U. S. A. 112 (2015) 1779–1784. Mallucci, S.H. Loh, L.M. Martins, Mitofusin-mediated ER stress triggers
[154] E.S. Dremina, V.S. Sharov, K. Kumar, A. Zaidi, E.K. Michaelis, C. Schoneich, neurodegeneration in pink1/parkin models of Parkinson’s disease, Cell.
Anti-apoptotic protein Bcl-2 interacts with and destabilizes the Death. Dis. 7 (2016) e2271.
sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA), Biochem. J. 383 [177] T. Cali, D. Ottolini, A. Negro, M. Brini, Alpha-synuclein controls mitochondrial
(2004) 361–370. calcium homeostasis by enhancing endoplasmic reticulum-mitochondria
[155] M.T. Khan, L. Wagner 2nd, D.I. Yule, C. Bhanumathy, S.K. Joseph, Akt kinase interactions, J. Biol. Chem. 287 (2012) 17914–17929.
phosphorylation of inositol 1,4,5-trisphosphate receptors, J. Biol. Chem. 281 [178] C. Guardia-Laguarta, E. Area-Gomez, C. Rub, Y. Liu, J. Magrane, D. Becker, W.
(2006) 3731–3737. Voos, E.A. Schon, S. Przedborski, alpha-Synuclein is localized to
[156] S. Marchi, A. Rimessi, C. Giorgi, C. Baldini, L. Ferroni, R. Rizzuto, P. Pinton, Akt mitochondria-associated ER membranes, J. Neurosci. 34 (2014) 249–259.
kinase reducing endoplasmic reticulum Ca2+ release protects cells from [179] D. Ottolini, T. Cali, A. Negro, M. Brini, The Parkinson disease-related protein
Ca2+-dependent apoptotic stimuli, Biochem. Biophys. Res. Commun. 375 DJ-1 counteracts mitochondrial impairment induced by the tumour
(2008) 501–505. suppressor protein p53 by enhancing endoplasmic reticulum-mitochondria
[157] C. Giorgi, K. Ito, H.K. Lin, C. Santangelo, M.R. Wieckowski, M. Lebiedzinska, A. tethering, Hum. Mol. Genet. 22 (2013) 2152–2168.
Bononi, M. Bonora, J. Duszynski, R. Bernardi, R. Rizzuto, C. Tacchetti, P. [180] T. Cali, D. Ottolini, M.E. Soriano, M. Brini, A new split-GFP-based probe
Pinton, P.P. Pandolfi, PML regulates apoptosis at endoplasmic reticulum by reveals DJ-1 translocation into the mitochondrial matrix to sustain ATP
modulating calcium release, Science 330 (2010) 1247–1251. synthesis upon nutrient deprivation, Hum. Mol. Genet. 24 (2015)
[158] S.C. Hedgepeth, M.I. Garcia, A.M. Wagner 2nd, S.V. Chintapalli, R.R. Snyder, 1045–1060.
G.D. Hankins, B.R. Henderson, K.M. Brodie, D.I. Yule, D.B. van Rossum, D. [181] S.C. Ling, M. Polymenidou, D.W. Cleveland, Converging mechanisms in ALS
Boehning, The BRCA1 tumor suppressor binds to inositol and FTD: disrupted RNA and protein homeostasis, Neuron 79 (2013)
1,4,5-trisphosphate receptors to stimulate apoptotic calcium release, J. Biol. 416–438.
Chem. 290 (2015) 7304–7313. [182] G. Manfredi, H. Kawamata, Mitochondria and endoplasmic reticulum
[159] M. Doghman-Bouguerra, V. Granatiero, S. Sbiera, I. Sbiera, S. Lacas-Gervais, crosstalk in amyotrophic lateral sclerosis, Neurobiol. Dis. 90 (2016) 35–42.
F. Brau, M. Fassnacht, R. Rizzuto, E. Lalli, FATE1 antagonizes calcium- and [183] J. Lautenschlager, T. Prell, J. Ruhmer, L. Weidemann, O.W. Witte, J.
drug-induced apoptosis by uncoupling ER and mitochondria, EMBO Rep. Grosskreutz, Overexpression of human mutated G93A SOD1 changes
(2016). dynamics of the ER mitochondria calcium cycle specifically in mouse
[160] C. Cardenas, M. Muller, A. McNeal, A. Lovy, F. Jana, G. Bustos, F. Urra, N. embryonic motor neurons, Exp. Neurol. 247 (2013) 91–100.
Smith, J. Molgo, J.A. Diehl, T.W. Ridky, J.K. Foskett, Selective vulnerability of [184] N. Bernard-Marissal, J.J. Medard, H. Azzedine, R. Chrast, Dysfunction in
cancer cells by inhibition of Ca(2+) transfer from endoplasmic reticulum to endoplasmic reticulum-mitochondria crosstalk underlies SIGMAR1 loss of
mitochondria, Cell Rep. 14 (2016) 2313–2324. function mediated motor neuron degeneration, Brain 138 (2015) 875–890.
[161] B. Schreiner, L. Hedskog, B. Wiehager, M. Ankarcrona, Amyloid-beta [185] E. Gregianin, G. Pallafacchina, S. Zanin, V. Crippa, P. Rusmini, A. Poletti, M.
peptides are generated in mitochondria-associated endoplasmic reticulum Fang, Z. Li, L. Diano, A. Petrucci, L. Lispi, T. Cavallaro, G.M. Fabrizi, M. Muglia,
membranes, J. Alzheimers Dis. 43 (2015) 369–374. F. Boaretto, A. Vettori, R. Rizzuto, M.L. Mostacciuolo, G. Vazza,
[162] E. Area-Gomez, M. Del Carmen Lara Castillo, M.D. Tambini, C. Loss-of-function mutations in the SIGMAR1 gene cause distal hereditary
Guardia-Laguarta, A.J. de Groof, M. Madra, J. Ikenouchi, M. Umeda, T.D. Bird, motor neuropathy by impairing ER-mitochondria tethering and Ca2+
S.L. Sturley, E.A. Schon, Upregulated function of mitochondria-associated ER signalling, Hum. Mol. Genet. (2016).
membranes in Alzheimer disease, EMBO J. 31 (2012) 4106–4123.

Das könnte Ihnen auch gefallen