You are on page 1of 21

REVIEWS

The cell biology of mitochondrial


membrane dynamics
Marta Giacomello1, Aswin Pyakurel1,2, Christina Glytsou1,2 and Luca Scorrano   1,2 ✉
Abstract | Owing to their ability to efficiently generate ATP required to sustain normal cell function,
mitochondria are often considered the ‘powerhouses of the cell’. However, our understanding of
the role of mitochondria in cell biology recently expanded when we recognized that they are key
platforms for a plethora of cell signalling cascades. This functional versatility is tightly coupled
to constant reshaping of the cellular mitochondrial network in a series of processes, collectively
referred to as mitochondrial membrane dynamics and involving organelle fusion and fission
(division) as well as ultrastructural remodelling of the membrane. Accordingly , mitochondrial
dynamics influence and often orchestrate not only metabolism but also complex cell signalling
events, such as those involved in regulating cell pluripotency , division, differentiation, senescence
and death. Reciprocally , mitochondrial membrane dynamics are extensively regulated by post-
translational modifications of its machinery and by the formation of membrane contact sites
between mitochondria and other organelles, both of which have the capacity to integrate inputs
from various pathways. Here, we discuss mitochondrial membrane dynamics and their regulation
and describe how bioenergetics and cellular signalling are linked to these dynamic changes of
mitochondrial morphology.

Krebs cycle
After the discovery of the Krebs cycle in the 1950s, and are transmitted into mitochondria. Moreover, the
A series of chemical reactions mitochondria were primarily associated with cellular OMM forms interfaces with other subcellular compart-
catalysed by enzymes that, bioenergetics. This view changed in the 1990s when ments — including prominently the endoplasmic reticu-
on oxidation of the acetyl mitochondria were shown to be important contribu- lum (ER), but also lysosomes, peroxisomes, endosomes,
portion of acetyl coenzyme A,
tors to apoptotic cell death1. These discoveries paved melanosomes, lipid droplets and the plasma membrane
generate reducing equivalents
(NADH and FADH2). the way to the idea that these organelles have a role in — to establish membrane contact sites (Fig. 1c).
cell signalling, as now corroborated by ample evidence. The multifaceted involvement of mitochondria in cell
Membrane contact sites Accordingly, mitochondria have been implicated in biology is accompanied by their high degree of morpho-
Points at which two biological
several complex cellular processes beyond cell death2,3, logical variability. Since early in mitochondrial research,
membranes run in parallel, at a
constant distance, for several
ranging from autophagy4,5 to stem cell differentiation6,7 it has been well established that mitochondria can come
nanometres. and regulation of immune response8 (Fig. 1a). in different shapes, with both their overall (length,
The twofold role of mitochondria as cellular pow- width, roundness) and their ultrastructural (organi-
erhouses and signalling organelles is paralleled by the zation of the mitochondrial membranes) morphology
fact that they are surrounded by two membranes: an showing large variation between cells, during cell cycle
inner mitochondrial membrane (IMM) and an outer and on metabolic or cellular signals. The name of these
mitochondrial membrane (OMM), characterized by organelles, which was coined by Carl Benda in 1898,
different composition and function (Fig. 1b). The IMM is the combination of the Greek words for ‘thread’ and
delimits the mitochondrial lumen (matrix) and can be ‘grain’9. Indeed, mitochondria can be found as isolated
further divided into two subcompartments: the inner organelles or joined to form larger networks; they can
1
Department of Biology, boundary membrane, running parallel to the OMM, and also be distributed unevenly (through regulated mito-
University of Padua,
the cristae — deeply convoluted, pleomorphic invagi- chondrial transport and positioning) in the cytosol to
Padua, Italy.
nations that provide the expansion of the surface area match local energy demands of the cell10. This changea-
2
Veneto Institute of Molecular
Medicine, Padua, Italy.
and harbour the machinery required for mitochondrial ble and adaptable nature of mitochondria involving their
✉e-mail: luca.scorrano@ respiration (Fig. 1b). By contrast, the OMM is smooth morphology and subcellular distribution is collectively
unipd.it and generally permeable (it only limits diffusion of mol- known as mitochondrial dynamics. Here, we focus spe-
https://doi.org/10.1038/ ecules bigger than ~5,000 Da) and behaves as a platform cifically on mitochondrial membrane dynamics and
s41580-020-0210-7 where cell signalling pathways converge, are decoded remodelling of mitochondrial morphology; for a review

Nature Reviews | Molecular Cell Biology


Reviews

a b
Autophagy Crista Crista IMM (IBM) ATP K+
Cell death junction channel MCU
Inflammation and Differentiation OMM IMS ATP
senescence synthase
Mitochondrion
K+
OPA1
Metabolism Stem cell Respiratory
maintenance chain Ca2+
Innate immune Migration complexes
response
Mitofusin Metabolites/
Matrix IMS proteins

c Plasma membrane
Melanosome
ER
Solute carriers/
MICOS Cytochrome c channels,
metabolite
exchangers
Lysosome

Mitochondrion d Basal condition

Fission Fusion

Lipid
droplet IMS MDC
Endosome protein
Peroxisome MDVs

Misfolded • Clearance of damaged • Increased ATP production


Lysosome mitochondrial mitochondria (mitophagy) • Exchange of matrix
Peroxisome membrane • ROS production content
protein • Mitochondrial distribution

Fig. 1 | Mitochondrial structure and function. a | Mitochondria coordinate protein complexes, dynamin-related protein optic atrophy protein 1 (OPA1),
various cellular functions. Key processes dependent on mitochondria are which forms oligomers at cristae junctions, disruption of which is associated
listed. b | Mitochondrial structure. The mitochondrial lumen (named ‘matrix’, with cristae opening and ATP synthase dimers, which were shown to
which is the site of the Krebs cycle, mitochondrial DNA replication, protein oligomerize into long double rows running along the cristae ridges.
biosynthesis) is surrounded by two membranes: the outer mitochondrial c | The OMM establishes networks of interactions with other organelles,
membrane (OMM) and the inner mitochondrial membrane (IMM). The OMM including membrane contact sites with the endoplasmic reticulum (ER),
acts as a diffusion barrier and also mediates the transduction of signals into plasma membrane, lysosomes and endosomes, lipid droplets, peroxisomes
and out of mitochondria. The IMM includes two main subcompartments: and melanosomes. These membrane contacts sites regulate mitochondrial
the inner boundary membrane (IBM) and mitochondrial cristae. The IBM functions. In addition, mitochondria release mitochondrial-derived vesicles
runs parallel to the OMM and hosts various channel transporters that (MDVs) and mitochondrial-derived compartments (MDCs), which
shuttle ions, ATP, ADP and small metabolites between the cytoplasm and the incorporate mitochondrial components. MDVs can communicate with
matrix. Mitochondrial cristae are membrane invaginations that serve as sites other organelles (lysosomes and peroxisomes), while MDCs are important
for oxidative phosphorylation (harbouring respiratory chain complexes and for mitochondrial quality control by removing misfolded proteins from
ATP synthase), mitochondrial DNA maintenance and iron–sulphur cluster mitochondrial membranes (Box 3). d | Mitochondrial shape is fundamental
biogenesis. Cristae are connected to the IBM via cristae junctions, which are for mitochondrial (metabolic) activity. For example, the efficiency of ATP
narrow clefts that close the cristae and thereby prevent the contents of the production is increased and the exchange of matrix content is favoured in
cristae from being released into the intermembrane space (IMS). Shaping fused organelles. By contrast, fragmented organelles produce more reactive
the IMM into cristae depends on various membrane-shaping proteins, oxygen species (ROS) and are efficiently cleared by mitophagy. However,
including the mitochondrial contact site and cristae organizing system mitochondrial fragmentation is also required for the equivalent distribution
(MICOS), which localizes to cristae junctions and also establishes contacts of mitochondria to daughter cells on cell division. MCU, mitochondrial
between the IMM and the OMM by interacting with outer membrane Ca2+ uniporter.

of mitochondrial transport and subcellular distribution, the cellular context have at least two far-reaching implica-
the reader is referred to ref.11. tions. First, they suggest that mitochondrial morphology/
The two main mechanisms of mitochondrial mem- shape (these two terms are used interchangeably to
brane dynamics are fusion and fission (division). refer to the visual characteristics of these organelles)
Another important aspect of mitochondrial membrane can be dynamically modulated, not only by the struc-
dynamics encompasses biogenesis and remodelling of tural components (that is, the lipids and proteins that
mitochondrial cristae. The experimental observations determine the structure of mitochondrial membranes)
that mitochondrial appearance changes depending on but also by signalling pathways. Second, they also

www.nature.com/nrm
Reviews

Table 1 | Human diseases caused by mutated fission/fusion factors Inner mitochondrial membrane. The cristae of the
IMM are the principal site of oxidative phosphorylation
gene Disease refs (OXPHOS), since they host all the complexes implicated
DRP1 Encephalopathy 264
in mitochondrial respiration, including the mitochondrial
Autosomal dominant optic atrophy 265 respiratory chain and the F1Fo-ATP synthase13,14. Furthermore,
cytochrome c, the only soluble OXPHOS member, local-
Microcephaly and pain insensitivity 266
izes mainly in the intracristal compartment3,15. Hence, the
GDAP1 Charcot–Marie–Tooth disease, axonal, type 2K , recessive 267
primary function of the IMM is bioenergetics, whereby
intermediate A , type 4A
the free energy stored in the reducing equivalents produced
INF2 Charcot–Marie–Tooth disease, dominant intermediate E 268
in the Krebs cycle is converted into ATP (Supplementary
Glomerulosclerosis, focal segmental, 5 269 Box 1). In addition, the ‘neck’ of cristae, called a cristae
MFF Leigh-like encephalopathy , optic atrophy and peripheral neuropathy 270 junction, can confine metabolites, proteins (such as
cytochrome c but also others) and solutes within the
Encephalopathy due to defective mitochondrial and peroxisomal 271

fission
cristae lumen (Fig. 1b).
We identify two main functional consequences of the
MFN2 Charcot–Marie–Tooth disease type 2A , hereditary motor and sensory 64
extreme compartmentalization of the IMM. The first
neuropathy VIA
impacts on bioenergetics. Cristae organization ensures
OPA1 Optic atrophy 1 66,67
the optimal conditions for ATP production, minimiz-
Behr syndrome 272
ing the diffusion of metabolites, protons and ADP
Mitochondrial DNA depletion syndrome 273 during respiration16. Super-resolution imaging offers
a formal proof to this concept, identifying individual
Syndromic Parkinson disease and dementia 274
cristae with different magnitudes of transmembrane
potential17. Accordingly, on activation of mitochondrial
suggest that mitochondrial morphology participates in respiration, cristae tighten, a process that can enhance
mitochondrial adaptation to specific cellular and tissue overall OXPHOS efficiency18–21. The second functional
demands, and governs mitochondrial response to cyto- consequence impacts on apoptosis. Because during
solic signals: for example, mitochondrial morphological apoptosis cytochrome c is required in the cytosol for
changes are not only the consequence of the recruitment the activation of effector caspases3, it must be released
of mitochondria in the cell death pathways but occur, from mitochondria. During cell death, cristae there-
for example, in response to nutrient stress, when mito- fore undergo a process of dramatic structural changes,
chondria first elongate and then fragment12 (Fig. 1d). referred to as ‘cristae remodelling’, that supports the
It is thus not surprising that deregulation of mitochon- intramitochondrial redistribution of cytochrome c from
drial dynamics (and in particular of fusion and fission) the cristae to the intermembrane space, followed by its
has been linked to several genetic disorders in humans complete release to the cytosol through apoptotic pores
(Table 1) and that the molecular effectors of these pro- in the OMM. Therefore, IMM organization into cristae
cesses are subject to various post-translational mod- needs to be tightly controlled. Mechanisms involved in
ifications (PTMs; Table 2), which allow the coupling regulating cristae organization will be discussed in the
of mitochondrial membrane dynamics to cell signal- following section.
ling, reflecting the sensitivity and responsiveness of
mitochondrial dynamics to cellular states. Outer mitochondrial membrane. Originally the barrier
Oxidative phosphorylation In this Review, we discuss the key mechanisms of between the invading endosymbiont and the hostile
(OXPHOS). A process involving
coupling between mitochondrial
mitochondrial membrane dynamics, focusing on our cytosol environment of the host cell, the OMM evolved
respiration and ATP production. progress in understanding how these dynamics link into a platform for signalling events that use mitochon-
to cell function. We specifically highlight the interplay dria. The OMM hosts proteins that are fundamental for
Mitochondrial respiratory between mitochondrial membrane dynamics and cell the organelle (and hence cell) physiology. They include
chain
signalling as well as the connectivity of mitochondria translocases mediating the transport of mitochondrial
An ensemble of protein
complexes whose coordinated to other organelles via membrane contact sites. At the precursor proteins; proteins that physically connect
activity catalyses the oxidation end of this Review, we hope that the reader will under- mitochondria with other subcellular compartments in
of reducing equivalents using stand that mitochondrial morphology is influenced by specific sites known as membrane contact sites; and
oxygen as a terminal electron the effect of cellular signals impinging on mitochondrial proteins that coordinate signal transduction pathways
acceptor.
dynamics and by the close connection between mito- with mitochondrial function. Finally, the OMM is cru-
F1Fo-ATP synthase chondria and other organelles, and reciprocally that cial for mitochondrial dynamics because it hosts all the
Protein complex located at the mitochondrial morphology influences not only mito- molecules involved in mitochondrial fusion and fission
inner mitochondrial membrane chondrial bioenergetics but also the metabolic pathways (see later).
which mediates Pi addition
and signalling cascades converging on mitochondria. The list of signalling cascades that use the OMM as
to ADP to generate ATP in a
process driven by the passage a platform grew to the point that mitochondria are now
of protons. Mitochondrial membranes considered ‘signalling organelles’, and this list is exten-
Mitochondria are surrounded by two structurally and sively covered by dedicated reviews22,23. For the scope
Reducing equivalents functionally different membranes. While the IMM is of this Review, it is interesting to note that in several
Electron donors in a redox
reaction, which become
mostly implicated in mitochondrial energy conversion, circumstances these signalling cascades are controlled
oxidized. In mitochondria, the OMM is the principal platform for mitochondrial by, or influence, mitochondrial dynamics. To give the
NADH and FADH2. signalling. reader a sense of the extent of this reciprocal influence,

Nature Reviews | Molecular Cell Biology


Reviews

Table 2 | Key post-translational modifications of mitochondrial dynamics proteins


Protein Modification Biological effect refs
DRP1 Phosphorylation (Ser585 and Ser616) by Promotes mitochondrial fission in mitotic cells 198

CDK1–cyclin B
Phosphorylation (Ser637) by PKA Inhibits fission by reducing DRP1 GTPase activity 104

Phosphorylation (Ser 600) by CaMKIα Activates fission 198

Dephosphorylation by calcineurin Activates fission 105

Sumoylation (Lys532, Lys535, Lys558, Lys568, Positively regulates mitochondrial fission 139,275

Lys594, Lys606, Lys608) by MAPL


S-Nitrosylation (Cys644) Increases DRP1 dimerization and mitochondrial 145

fission; causes neuronal damage


Ubiquitylation by APC/C components Targets DRP1 for proteasomal degradation, allowing 204

mitochondrial elongation after release from mitotic


arrest
Ubiquitylation by MARCH5 Targets DRP1 for proteasomal degradation; 276

decreases mitochondrial fission


FIS1 Ubiquitylation by MARCH5 Targets FIS1 for proteasomal degradation; decreases 276

mitochondrial fission
GDAP1 Ubiquitylation by parkin (Lys50, Lys172, Targets GDAP1 for proteasomal degradation; 276

Lys173, Lys188, Lys190, Lys203, Lys206, enhances mitophagy


Lys207 , Lys214)
MFN1 Ubiquitylation by parkin Targets MFN1 for proteasomal degradation; 277

increases mitochondrial fission, activates mitophagy


Ubiquitylation by MARCH5 Favours fission 278

Phosphorylation by ERK (Thr526) Favours fission 279

Phosphorylation by PKC isoform-β2 (Ser86) Inhibition of mitochondrial fusion during ischaemia– 158

reperfusion injury
MFN2 Phosphorylation by PINK1 (Thr111, Ser442, Favours interaction of MFN2 with parkin; promotes 187

Tyr448) fission and mitophagy


Phosphorylation by PKA (Ser442) Affects vascular muscle cell proliferation 280

Ubiquitylation by parkin Regulates MFN2 levels and mitochondrial fusion 181

Ubiquitylation by MARCH5 Targets MFN2 for proteasomal degradation; inhibits 281

fusion
Phosphorylation by JNK (Ser27) Promotes recruitment of the ubiquitin ligase 159

HUWE1, leading to MFN2 ubiquitylation and


proteasomal degradation; inhibits mitochondrial
fusion
OPA1 Cleavage by OMA1 Inhibits mitochondrial fusion 81

Cleavage by YME1L Required for mitochondrial fusion 282

Proteolysis by PARL Required to generate a pool of short OPA1 soluble in 82

the IMS and to form the OPA1 oligomer that inhibits


cristae remodelling
Acetylation (Lys936, Lys961) Lowers OPA1 GTPase activity 173

O-linked β-N-acetylglucosaminylation Increases mitochondrial fragmentation 175

(undefined Ser/Thr residues)


APC/C, anaphase-promoting complex/cyclosome; CaMKIα, calcium/calmodulin-dependent protein kinase Iα; CDK1,
cyclin-dependent kinase 1; DRP1, dynamin-related protein 1; ERK , extracellular-signal-regulated kinase; FIS1, mitochondrial
fission 1 protein; GDAP1, ganglioside-induced differentiation-associated protein 1; JNK , JUN amino-terminal kinase; MARCH5,
membrane-associated RING-CH protein 5; MAPL , mitochondrial-anchored protein ligase; MFN, mitofusin; OPA1, optic atrophy
protein 1; PARL , presenilin-associated rhomboid-like protein; PINK1, PTEN-induced kinase 1; PKA , protein kinase A ; PKC, protein
kinase C; YME1L , YME1-like ATPase.

we decided to illustrate here only three examples and is controlled by mitochondrial dynamics. The role
BCL-2 family of bidirectional mitochondrial dynamics–signalling of mitochondria in apoptosis was identified when the
A group of evolutionarily cascade crosstalk. antiapoptotic protein BCL-2 was found associated with
conserved proteins that Apoptosis, the stereotypical, genetically controlled the OMM24. Here, together with other members of the
harbour a BCL-2 homology
process of programmed cell death, is the paradigmatic BCL-2 family, BCL-2 controls OMM permeabilization
domain. Mostly known for their
regulatory role in programmed example of a cellular signalling cascade that requires the that is accompanied by mitochondrial fragmentation
cell death. OMM, leads to mitochondrial morphological changes and cytochrome c release25. Apart from regulating the

www.nature.com/nrm
Reviews

Nucleoids formation of apoptotic pores, BCL-2 family members oligomerization37,38. MIC60 is the largest and a core
The literal meaning of this can interact with and influence the activity of key MICOS complex component39,40. The MICOS complex
term is ‘nucleus-like’; it refers mitochondrial fusion and fission proteins26–28. disassembles in MIC60-deficient mitochondria of yeast
to a cluster of proteins and As another example, several A-kinase anchoring and human cells, where cristae disconnect from the
mitochondrial DNA, not
surrounded by a membrane.
proteins, which recruit cAMP-dependent serine/threonine- inner boundary membrane. In addition, these cells are
specific protein kinase A (PKA) are associated with the characterized by OXPHOS defects, abnormal distribu-
OMM, thus regulating cAMP signalling in time and tion of nucleoids and impaired mitochondrial translation.
space29. This OMM association also leads to discrete The conserved function of MIC60 on cristae formation
areas of increased PKA-dependent phosphorylation at the and/or maintenance of cristae junctions39–41 was fur-
OMM30, influencing the activity of several mitochondrial ther reinforced by elegant experiments whereby fungal
dynamics proteins as we will see in detail later. Mic60 was shown to induce membrane deformation
Finally, OMM localization of mitochondrial in liposomes in vitro and it was sufficient to cause invag-
antiviral-signalling protein (MAVS) is essential to mount inations that resemble nascent cristae in bacterial mem-
a response to viral infection. MAVS also regulates the branes following ectopic expression42,43. However, these
activity of the mitochondrial fusion protein mitofusin 1 invaginations are not fully formed cristae, raising the
(MFN1; see also later) to promote fusion, which supports question of how a completely formed cristae structure is
MAVS-mediated signal transduction8. achieved in mitochondria of higher organisms. One pos-
In addition to regulating signalling events at the sibility is that MIC60 cooperates with other proteins that
OMM, these three signalling cascades extend beyond shape cristae architecture. For example, MIC60 can inter-
the OMM: BCL-2 family members also induce cristae act with two other proteins that repeatedly affect cristae
remodelling3; similarly, one of the PKA substrates is a shape (see also later), F1Fo-ATP synthase via MIC27 (ref.44)
cristae biogenesis factor, MIC19 (ref.31) (see the next and the dynamin-related protein (Box 1) OPA1 (Mgm1p
section); MAVS is involved in regulating mitochondria– in yeast) via MIC19 (ref.45). While the bacterial membrane
ER contacts32. Overall, the OMM can be truly regarded harbours the ATP synthase, a classical dynamin-related
as the platform of mitochondrial integration within protein like OPA is not found in prokaryotes, suggesting
cellular signalling. that perhaps the OPA1–MIC60 interaction occurred later
in evolution and could have been the driving force for
Mitochondrial cristae dynamics cristae shaping.
Mitochondrial cristae are dynamic structures that adapt At the cristae edge, F1Fo-ATP synthase is organized
their architecture in response to physiological inputs and in dimers, which can further assemble into oligomers.
in pathology, notably apoptosis. The organization of the The long double rows of ATP synthase dimers along the
IMM into cristae, also known as cristae biogenesis, is a cristae ridges have been observed across species46,47. ATP
complex process with multiple players. First, it can be synthase dimers have been proposed as crucial drivers
intrinsically caused by spontaneous forces generated of cristae morphology, because their formation can bend
by lipid- and protein-driven membrane curvature33. lipid bilayers. In turn, the elastic membrane conforma-
In addition, the OMM likely serves as a border to limit tion favours their association into rows. In line with this
the extension of cristae structures, while the protein-rich idea, deletion of the ATP synthase dimerization sub­
matrix determines the minimum volume of the inner units causes loss of invaginations, with the IMM being
compartments of the organelles. These ‘spontaneous’ inflated into an onion-like or balloon-like shape46,48.
organizers of cristae shape work in concert with specific Moreover, ATP synthase inhibitor factor 1 (IF1) neg-
protein modulators of cristae architecture. atively impacts ATP synthase oligomerization, at the
A key multiprotein complex regulating cristae bio- same time disturbing cristae organization and reduc-
genesis is the mitochondrial contact site and cristae ing their density during physiological and pathological
organizing system (MICOS). This large heterooligomeric conditions49. It should be noted, however, that mutated
protein structure of the IMM is composed of MIC10, ATP synthase dimerization subunits lead to altered
MIC12, MIC19 MIC25, MIC26, MIC27 (also known as IMM dynamics, impaired OXPHOS, membrane poten-
MOMA1 and APOOL) and MIC60 (also known as mito- tial reduction and disruption of lipid microdomains50.
filin), organized in two independent protein assemblies, Thus, whether ATP synthase assemblies directly impact
MIC27–MIC10–MIC12 and MIC60–MIC19, linked on cristae curvature is still unclear.
via MIC19 (ref.34). The composition of MICOS is not Another key regulator of cristae architecture is
still completely defined. For example, QIL1 (encoded OPA1 (refs51,52), which is important for cristae junc-
by C19orf70) was recently identified in human cells as tion formation and maintenance, and also, as will be
an essential MICOS component35. A strong hint that discussed in the following section, independently reg-
MICOS is associated with cristae morphogenesis comes ulates mitochondrial fusion. In yeast and mammalian
from evolutionary analyses indicating that MICOS cells, Mgm1/OPA1 ablation or alteration leads to cris-
components are found exclusively in organisms with tae disorganization and widening of cristae junctions.
cristae-containing mitochondria36. Biochemically, OPA1-driven cristae biogenesis corre-
Functionally, each MICOS component exerts a lates with the stability of high molecular weight com-
specific effect on membrane shape, and together they plexes of OPA1 (notably, these complexes are disrupted
can generate the curvature changes required for cris- on apoptotic stimulation, which drives apoptotic IMM
tae biogenesis. MIC10 forms a hairpin in the mem- remodelling, facilitating cytochrome c release; see also
brane and harbours a glycine-rich motif required for later). Accurate measurements indicate that the stability

Nature Reviews | Molecular Cell Biology


Reviews

Box 1 | Membrane shaping by dynamin-related proteins bridge between unprocessed, membrane-bound OPA1
(so-called long OPA1 (L-OPA1)) molecules (see also the
Dynamin-related proteins (DrPs) are a group of GtPases whose main function is to next section).
shape biological membranes. Members of the DrP family include dynamins, dynamin- Proteomic analysis further identified MIC60 as well
like proteins, Mx dynamin-like GtPases, optic atrophy protein 1 (OPa1), mitofusin1
as multiple ATP synthase subunits as components of
(MFN1), MFN2 and guanylate-binding proteins229.
the OPA1 high molecular weight complexes associ-
DrPs undergo GtP-dependent conformational changes that drive their oligomerization
and endow them with the capability to induce membrane fission. their structure includes ated with cristae remodelling45,53,54, raising the question
a GtPase domain and a GtPase effector (GeD) domain, which together form a flexible of whether OPA1 controls cristae shape alone or with
intramolecular unit named ‘bundle signalling element’. rearrangement of this element these other players. Formal epistatic analyses revealed
promotes nucleotide-dependent GtPase domain dimerization, translating GtP binding that in mammals OPA1 lies upstream of both MIC60
into self-assembly of the DrPs into spirals/rings229. these conformational changes favour and ATP synthase dimerization in the regulation of
GtP hydrolysis, which in turn narrows the adjacent dynamin rings, induces DrP rotation cristae shape53,54. Similarly, in yeast lacking Mgm1, ATP
and membrane constriction up to the formation of a hemifusion membrane that synthase oligomerization is impaired52,57,58, lending
undergoes scission230. further support to the central role of OPA1 in sculpt-
One of the best studied DrPs is dynamin 1, which is essential for endocytosis. ing cristae. However, this model lacks formal proof,
this is a stepwise process that leads to the formation of omega-shaped invaginations
for example, in heterologous models such as bacteria.
underneath the plasma membrane, which are ultimately released into the cytoplasm
as excised vesicles. endocytosis starts on interaction of adaptor proteins with Lastly, there is evidence that cristae remodelling via
transmembrane proteins protruding from the lipid bilayer towards the cytosol. OPA1 is coupled to the formation of ER–mitochon-
These complexes form scaffolds for the recruitment of clathrin triskelion units at the dria membrane contact sites, whereby signals from the
neck of endocytic vesicles, and then trigger membrane deflection up to the formation OMM are relayed to OPA1, inducing its proteolytic pro-
of coated pits engulfing a molecular cargo, and provide an anchor for Bin/amphiphysin/ cessing, via the OMM protein mitofusin 2 (MFN2; see
rvs (Bar) motif-containing proteins such as endophilin and amphiphysin231. also later).
The synergistic activity of these proteins controls membrane remodelling by supporting Finally, it is conceivable that other factors partici-
dynamin 1 oligomerization into α-helices and twisting of its GtPase domain, followed pate in cristae biogenesis and maintenance. For exam-
by dynamin 1 oligomers wrapping around budding endocytic vesicles. these events
ple, protein FAM92A1, which carries a Bin/amphiphysin/
result in GtP cycle-dependent membrane fission229,232–234. this mechanism of membrane
Rvs (BAR) domain and resides at the matrix side of the
fission is conserved between DrPs, no matter whether it occurs on the plasma or
intracellular membranes. IMM, can generate membrane curvature through pref-
the activity of DrPs that act on intracellular membranes is not restricted to fission, erential interaction with negatively charged phospholip-
with some DrPs facilitating fusion and tubulation (that is, generation of tubular ids such as phosphatidylinositol 4,5-bisphosphate and
invaginations or projections). through these activities, DrPs control the morphology cardiolipin and hence orchestrate cristae shape59. The
of different organelles. sometimes various DrPs act to remodel the same organelle, as relationships between this and the other newly discov-
occurs for DrP1, OPa1 and mitofusins in mitochondria. alternatively, a single protein ered cristae shape factors and MICOS, ATP synthase and
can act on two organelles. For example, atlastin 1 is necessary for the morphogenesis OPA1 are unclear and warrant further investigation.
of both the Golgi apparatus and the endoplasmic reticulum, while DrP1 shapes both
peroxisomes and mitochondria235,236. Mitochondrial fusion and fission
DrP-mediated membrane remodelling impacts on the function of individual
Fusion and fission are key events regulating mitochon-
organelles, on their interconnection and on cell physiology and fate. in light of these
different functional consequences, mutations in the genes encoding DrPs have been drial morphology that need to be balanced to support
associated with various human disorders. For instance, mutations in SPG3A, encoding normal mitochondrial function and prevent disease60.
atlastin 1 (ref.237), cause hereditary spastic paraplegia. as another example, mutated Accordingly, several human diseases have been associ-
forms of the endoplasmic reticulum-shaping protein dynamin 2 have been associated ated with mutations in the regulators of these dynam-
with inherited peripheral neuropathy (Charcot–Marie–tooth disease)238. Charcot– ics (Table 1). Several excellent reviews summarize our
Marie–tooth disease-causing mutants affect dynamin 2 binding to membranes or current understanding of mitochondrial fission and
membrane curvature generation, ultimately altering its fission-related functions. fusion61–63. Here, we provide the reader with the basic
Diseases associated with mutations in the genes encoding DrP1, mitofusins and OPa1 knowledge of these processes, which is needed to under-
are shown in Table 1. stand how signalling pathways can impinge on mito-
chondrial morphology and how mitochondrial form
of these complexes directly correlates with the cristae and function are linked.
junction diameter and cristae lumen width; similarly,
OPA1 overexpression decreases cristae lumen width, Mitochondrial fusion. In mammalian cells, the fusion of
tightening the cristae53,54. Mechanistically, one study mitochondria is regulated by two mitofusins — proteins
showed that Mgm1 can form helical filaments on the belonging to the dynamin-related family of large
inside of membrane tubes with the shape and dimen- GTPases (Box 1): MFN1 and MFN2 (the encoding gene
sions of cristae junctions. They further suggested that a is mutated in Charcot–Marie–Tooth disease type 2A)64
dynamin-like power stroke (Box 1) in such a left-handed located in the OMM (of note, there is only a single
helical assembly on the inside of a membrane tube mitofusin in yeast, Fzo1p)65 and OPA1 (mutations of
would constrict its diameter55. Cristae formation and the encoding gene are associated with dominant optic
Bin/amphiphysin/Rvs (BAR) stability can also be explained by homotypic interactions atrophy)66,67 located in the IMM68 (Fig. 2,Table 1).
domain between Mgm1/OPA1 on opposing membranes47,56. For MFN1 and MFN2 display a very high degree
Domains that mediate protein OPA1, it has been proposed that this tethering of mem- of homology and similar structural organization69.
dimerization, first discovered in
three protein classes named
branes and cristae maintenance involves a protease- However, genetic and biochemical studies highlighted
Bin, amphiphysin and Rvs cleaved, soluble OPA1 (so-called short OPA1 (S-OPA1)) that the two mitofusins display different functions:
(BAR). present in the intermembrane space that serves as a MFN1 is a core component of the fusion reaction

www.nature.com/nrm
Reviews

ER
(cross section)
INF2 Calcineurin-dependent
Actin dephosphorylation
OMM
Spire 1C
DRP1
Mitochondrion DRP1 Phosphorylation
Calcium

Calcineurin DRP1 receptor


ER
Ca2+ influx

Fusion Fission

MFN1

MFN1 Short OPA1 Cardiolipin


GTPase domain Long OPA1
Transmembrane domain OMM
HR2
OMM
HR1
IMM IMM Protease
Matrix
OPA1 proteolytic processing

Fig. 2 | The mitochondrial life cycle involving fusion and fission. Fission and fusion cooperate to regulate mitochondrial
morphology and in extension its function allowing the mitochondrial network to adapt to the needs of a cell and to
external cues (Fig. 1d). Fission is primarily orchestrated by dynamin-related protein 1 (DRP1). DRP1 is phosphorylated on
Ser637 by protein kinase A , which maintains DRP1 in the cytoplasm. Fission occurs when DRP1 is dephosphorylated by
calcineurin and recruited to the mitochondrial surface (where it binds to its receptors). This has been shown to occur at sites
where the endoplasmic reticulum (ER) wraps mitochondria. Here, DRP1 oligomerizes and induces GTP hydrolysis-mediated
membrane constriction. The activity of DRP1 is supported by actin polymerization at the ER–mitochondria interface
mediated by the actin-nucleating proteins inverted formin 2 (INF2) and formin-binding protein spire 1C. This actin
filament accumulation could drive initial mitochondrial constriction that supports subsequent DRP1 polymerization.
The ER–mitochondria contact sites are also a functional platform for the transmission of Ca2+ signals from the ER to
mitochondria, which supports cristae remodelling, amplifies cytochrome c release and fuels mitochondrial apoptosis (Fig. 3b).
Fusion relies on two GTPases residing at both the outer mitochondrial membrane (OMM) and the inner mitochondrial
membrane (IMM), mitofusin 1 (MFN1) and optic atrophy protein 1 (OPA1), respectively. Fusion starts with the docking of
two MFN1 molecules in trans, likely through the HR2 domain or GTPase domain; this association induces conformational
changes that drive GTP hydrolysis by MFN1 molecules, leading to fusion of the two OMMs. In the IMM, the unprocessed
OPA1 form (known as long OPA1) interacts with lipid cardiolipin in trans, whereby OPA1 is fusion competent.
This association is promoted in the presence of short OPA1, which is generated by proteolytic degradation of long
OPA1. Another mitofusin, MFN2, is also required for fusion, but its mechanism in this process is elusive (it is an established
tether between mitochondria and other organelles, prominently the ER (Box 2)).

together with OPA1, whereas the exact role of MFN2 a minimal MFN1 (formed by the predicted GTPase
in fusion remains elusive and it has been shown to par- domain and the distal part of the carboxy-terminal tail)
ticipate in mitochondrion–mitochondrion interaction, suggest that MFN1 functions as a tether between fusing
as well as in juxtaposition of mitochondria with other mitochondria. In support of the tethering role of MFN1,
organelles (in particular with the ER)68,70,71, which will the minimal MFN1 also dimerizes in a GTP-dependent
be further discussed later. The crystal structures of manner, leading to artificial membrane clustering72.

Nature Reviews | Molecular Cell Biology


Reviews

Rhomboid protease Whether the same is true for full-length MFN1 is still mitochondrial fusion77, is still unclear. Mammalian
Intramembrane serine to be determined. OPA1 might also be processed by the Rbd1p orthologue
proteases that share a The role of OPA1 in mitochondrial membrane presenilin-associated rhomboid-like protein (PARL;
common catalytic domain fusion was established in the early days of mitochondrial whether this processing occurs directly or indirectly is a
composed of six
membrane-spanning segments.
dynamics, and the function ascribed was coordination matter of debate)82–84. PARL-mediated processing is not
of OMM and IMM fusion. Around the same time, the essential for the generation of S-OPA1 but participates in
role of OPA1 in mitochondrial cristae remodelling was its further maturation. When PARL is deleted, S-OPA1
also discovered51,68, raising the question of how a single is still produced, but the fraction soluble in the inter-
protein can mediate these two distinct processes. The membrane space is lacking. This specific fraction of
elucidation of the crystal and cryo-electron microscopy S-OPA1 does not participate in mitochondrial fusion
(cryo-EM) structures of yeast Mgm1 now provides struc- but participates in the formation of the OPA1 high
tural support for its different functions. A key feature of molecular weight oligomers that maintain cristae junc-
the quaternary structure of Mgm1 is its organization in tions82. Overall, processing of OPA1 has extensive impact
right- or left-turned helical assemblies. Right-turned on mitochondrial dynamics, and how exactly different
assemblies at the inner boundary membrane can exert forms of OPA1 function awaits further refinement.
a pulling force on the membrane, eventually leading to
bending and fusion. Left-turned assemblies inside the Mitochondrial fission. The fission of mitochondria
cristae can conversely constrict the membrane, thereby is primarily carried out by dynamin-related protein 1
diminishing the cristae lumen55. The possibility to adopt (DRP1; Dnm1 in yeast)85, which translocates from the
both turns is therefore an economical solution to a pro- cytosol to mitochondria, binding to its OMM partners
tein which participates in both fusion and cristae remod- (called receptors): mitochondrial fission factor (MFF),
elling. Whether the same organization is conserved for mitochondrial dynamics protein of 49 kDa (MID49),
OPA1 is unclear, especially because of the limited degree MID51 and mitochondrial fission 1 protein (FIS1)86–89
of homology between the two molecules. Another key (Fig.  2) . Following this binding, DRP1 oligomerizes
aspect of OPA1 function that remains unresolved despite and drives scission. Mechanistic insights into DRP1-
the elucidation of Mgm1 structure is how its direct mediated scission have been gained recently through
interaction with MFN1 (ref.56) can coordinate OMM cryo-EM analysis of MID49/MID51–DRP1–GTP
and IMM fusion68. A plausible model involves the coor- complexes. This analysis revealed that GTP binding to
dinated formation of OMM fusion pores mediated DRP1 induces formation of linear polymers on the mito-
by GTP hydrolysis-stimulated MFN1 oligomerization73 chondrial membrane stabilized by the interaction with
followed by IMM fusion by right-turned helical assem- the receptors. Subsequent GTP hydrolysis was associa­
blies of OPA1, perhaps somehow activated by MFN1. ted with polymer shortening while curling into closed
Finally, fusion depends on the appropriate balance rings with an inner diameter of 16 nm, well in the range
between L-OPA1 and S-OPA1. In vitro studies revealed of complete mitochondrial constriction90.
that L-OPA1 establishes two types of interactions between In yeast, Fis1 is mainly engaged to recruit Dnm1 to
membranes: homotypic trans interaction between two the mitochondrial membrane (of note, yeast Fis1 does
L-OPA1 molecules that are fusion incompetent; and not directly bind Dnm1 but acts as a membrane adapter
hetero­typic trans interactions between L-OPA1 and cardio­ for additional mitochondrial division proteins Mdv1 and
lipin that are fusion competent (Fig. 2). Similarly to the Caf4, which do not have homologues in mammals)91–93.
role of S-OPA1 in cristae biogenesis, addition of S-OPA1 By contrast, mammalian mitochondria show a normal
to the trans L-OPA1–cardiolipin complex enhanced phenotype after FIS1 deletion86. Yet, FIS1 overexpres-
membrane fusion74. sion leads to mitochondrial fragmentation87 — an effect
In yeast, Mgm1 is proteolytically processed by a sin- that could be secondary to other FIS1 functions such as
gle enzyme: a mitochondrial rhomboid protease, Rbd1, regulation of mitochondrial Ca2+ fluxes and function94,
that generates a pool of short-form Mgm1 (refs75,76); this enhanced interaction with lysosomes (which can mark
processing as well as the presence of both long and short sites of mitochondrial fission)95 and inhibition of the
forms of Mgm1 was found to be required for regulation mitochondrial fusion machinery96. This latter effect,
of IMM structure and for mitochondrial fusion56,77, sub- however, remains contentious94,96, raising questions on
stantiating the role of Mgm1/OPA1 proteolytic process- its contribution to the mitochondrial fragmentation
ing in mitochondrial membrane dynamics. Mammalian observed on FIS1 overexpression. Instead, MFF seems
OPA1 is processed by multiple enzymes. The key enzyme to prevail over the other DRP1 receptors in mammals.
for generating S-OPA1 is YME1L1 (ref.78). In addition, However, it should be noted that the phenotype of
OPA1 is processed by the IMM peptidase OMA1, lead- the Mff-knockout mouse is much milder than that
ing to mitochondrial fission and fragmentation on mito- of the Drp1-knockout mouse (Drp1 is also known as
chondrial dysfunction79–81; these mechanisms might also Dnm1l)60,97. It could be that other DRP1 receptors can
be used during cell death (see the section Crosstalk with compensate for MFF absence98 or that the pathological
cell function). Whether S-OPA1 generated by OMA1- unbalance between fusion and fission is less pronounced
mediated processing has a direct profission role79 or in Mff-knockout mice as compared with Drp1-knockout
whether the changes in mitochondrial morphology mice60. There is also evidence that DRP1 oligomerizes
observed on OMA1 activation are caused by an imbal- even in the absence of DRP1 receptors, but in this case
ance between the L-OPA1 form and the S-OPA1 form, structural studies indicate nucleotide-independent con-
which, like for Mgm1, are both required for efficient striction of membranes to a final diameter of 30–70 nm

www.nature.com/nrm
Reviews

(ref.99),which is not sufficient to drive the complete also described) to physically link the two organelles.
mitochondrial constriction, suggesting a requirement Bridging of ER and mitochondria also can be mediated
for other DRPs (Box 1) to complete scission100 (although by the interaction of ER-resident vesicle-associated
the role of other DRPs beyond DRP1 in mitochondrial membrane protein-associated protein B (VAPB)
fission remains contentious)101,102. with OMM protein tyrosine phosphatase-interacting
As mentioned already, DRP1 is a cytosolic pro- protein 51 (PTPIP51) (Box 2). It has also been reported
tein that must translocate to mitochondria to induce that spatial dynamics of both organelles are coupled and
fission. This translocation depends on phosphorylation– depend on acetylated microtubules112.
dephosphorylation of Ser637 of DRP1. This modifica- As mentioned earlier, spatially, mitochondrial fis-
tion, which is mediated by PKA, decreases DRP1 GTPase sion occurs at sites of proximity to the smooth ER,
activity and leads to mitochondrial elongation due to which wraps around mitochondria (Fig.  2). At these
unopposed mitochondrial fusion103,104. The same site membrane contact sites, DRP1 cooperates with the
can be dephosphorylated by the Ca2+-dependent phos- actin-nucleating proteins inverted formin 2 (INF2) and
phatase calcineurin, which was shown to drive DRP1 formin-binding protein spire 1C, which causes actin
mitochondrial association and fission105. Although the accumulation that precedes DRP1 recruitment to sites
role of these phosphorylation–dephosphorylation events of fission. This actin filament accumulation could drive
in regulating mitochondrial fission was recently ques- initial mitochondrial constriction that supports subse-
tioned106, cryo-EM analysis of the MID49/MID51–DRP1 quent DRP1 polymerization113,114 (Fig. 2). These MERCs
complex predicts that Ser637 phosphorylation inhibits also allow the delivery of Ca2+ to mitochondria, which
the DRP1–receptor association90, indicating that dephos- activates IMM constriction in a DRP1-independent
phorylation is indeed required to translocate DRP1 to manner to complete fission115 and can also drive mito-
mitochondria (Fig. 2). chondrial apoptosis (see also the section Crosstalk
Finally, it is important to note that the process of with cell function). In addition to regulating fission,
mitochondrial fission is much more complex than DRP1- MERCs are important sites for the exchange of lipids
mediated membrane scission. As will be discussed in the between ER and mitochondria116. Loss of lipid homeo-
following section, mitochondrial fission sites are tightly stasis at MERCs was shown to lead to abnormal mito-
associated with ER tubules, which together with actin chondrial architecture, with markedly shortened cristae
cytoskeleton support membrane constriction (Fig. 2). and mitochondrial hyperfusion, leading to dysfunction of
mitochondrial respiration and premature senescence117
Interactions with other organelles (in accordance with the regulation of both processes by
In the context of mitochondrial membrane dynamics, mitochondrial dynamics; see the section Crosstalk with
it is important to consider that the OMM is engaged in cell function). We expect that further, in-depth character-
an extensive network of interactions with membranes ization of the ER–mitochondria interface will reveal novel
of other organelles establishing membrane contact sites. regulators of mitochondrial morphology and function, as
The notion that mitochondrial dynamics could be well as their interplay.
regulated by contact sites with other organelles stemmed
from two observations: that the fusion protein MFN2 Plasma membrane–mitochondria interactions. Beyond
also tethers mitochondria to the ER71, and, as mentioned MERCs, functional crosstalk between mitochondrial
earlier, that mitochondrial fission spatially localizes at membrane dynamics and other membranes is now start-
sites of proximity to the ER107. This organization of the ing to emerge. For example, in mammary stem cells, mito-
fission platforms by the ER is reverberated by the finding chondria can be tethered to the plasma membrane by the
that in mammalian cells the putative DRP1 receptor FIS1 binding of MFN1 to the plasma membrane-associated
regulates contacts with the ER108 and with lysosomes95, pool of protein kinase C isoform-ζ, which allows asym-
further highlighting the tight connection between mem- metric distribution of fused mitochondria to regulate
brane dynamics and interorganelle contact sites. We will stem cell fate decisions (see also the section Crosstalk
now provide an overview of the different interactions with cell function)118. Mitochondria–plasma membrane
between mitochondria and other organelles, how they contacts are also involved in the regulation of Ca2+ influx
are involved in regulation of mitochondrial dynamics from the extracellular medium, which is controlled by
and how this could impact on mitochondrial function. MFN2 (ref.119). It is important to note that while the
mitochondria–plasma membrane tethering machin-
ER–mitochondria interactions. The interaction ery is somewhat understood in yeast, where a handful
between mitochondria and the ER is to date the best of tethers have been identified120, it remains elusive in
studied type of a membrane contact site. This inter- mammalian cells. We expect that its characterization will
face was first discovered when ER membrane patches prompt the discovery of additional crossroads between
were biochemically isolated attached to the OMM109 mitochondrial shape and function and how they are
and named mitochondria-associated ER membranes. regulated by plasma membrane-derived signalling.
Mitochondria-associated ER membranes are tethered
Iron–sulphur clusters to mitochondria by protein bridges, establishing mito- Interactions between endosomes/lysosomes and mito-
Metal prosthetic groups chondria–ER contact sites (MERCs)110,111 (Box 2). A key chondria. Mitochondria are the site of biogenesis of
synthesized in mitochondria
and acting as cofactors to
MERC tether is MFN2, which by localizing at both iron–sulphur clusters, which requires a constant supply
catalyse several redox the ER and the OMM can homo-oligomerize (tether- of iron, which is taken up by endocytosis121,122. Super-
enzymatic activities. ing hetero-oligomers between MFN2 and MFN1 were resolution microscopy revealed interactions between

Nature Reviews | Molecular Cell Biology


Reviews

Box 2 | Mitochondrial juxtaposition with the er: a prototypical membrane contact site
Mitochondrial contacts with the endoplasmic reticulum (er) — mitochondria–er contact sites (MerCs) — are one
of the best studied membrane contact sites110, and several functions have been associated with them (see the figure).
the existence of MerCs has been evidenced by partial colocalization of fluorescently labelled mitochondria and er239,
isolation of er membranes linked to mitochondria (mitochondria-associated er membranes (MaMs))109 and the observation
of electron-dense, rod-shaped structures connecting the two organelles110,240,241.
For a long time, it has been known that MerCs host microdomains of high Ca2+ concentration. it is now established that
one of the key functions of MerCs is mediating Ca2+ transfer between the er and mitochondria. energy-dependent
mitochondrial Ca2+ uptake, described in the 1960s, was explained by Mitchell’s chemiosmotic theory192. substrate
oxidation drives Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCu), which sustains the activity of three matrix
dehydrogenases, connecting mitochondrial Ca2+ handling and physiology242. rapid entry occurs at sites of proximity
between mitochondria and the er. Ca2+ transfer is mediated by the inositol trisphosphate receptor (iP3r)–GrP75–
voltage dependent anion channel (vDaC) complex, while additional MaM-associated proteins, such as FuN14
domain-containing protein 1 (FuNDC1)243 and disrupted in schizophrenia 1 protein (DisC1) modulate Ca2+ uptake244.
However, the first MaM-associated protein was phosphofurin acidic cluster sorting protein 2 (PaCs2), which has been
shown to impact mitochondrial morphology and lipid synthesis245. indeed, enzymes fundamental for phospholipid
biosynthesis reside at MerCs, and conversion of phosphatidylserine into phosphatidylcholine requires the mutual
exchange of their intermediates246. another MerC protein is mitofusin 2 (MFN2), which resides both in the er and in the
outer mitochondrial membrane (OMM). er- and OMM-resident forms of MFN2 can oligomerize to physically link these
organelles71,247 in several tissues, from pro-opiomelanocortin (POMC) neurons, where MFN2-dependent MerCs control
leptin resistance221, to the heart, where MFN2 deficiency uncouples mitochondria from the sarcoplasmic reticulum,
resulting in impaired bioenergetics248, to glioma stem cells, where it regulates glycan levels249. this suggests that MFN2
acts as a bona fide tether250. Nevertheless, two independent reports concluded that MFN2 deletion increases er–
mitochondria tethering251,252. MFN2 domain organization resembles that of other proteins of the dynamin superfamily
involved in membrane tethering. it is therefore difficult to rationalize a direct antagonist of this function. Perhaps, under
the conditions studied in these reports, the increased tethering is due to compensatory effects mediated by, for example,
er stress253, or increased expression of other tethers. MerCs can be also tethered by the interaction of er-resident
vesicle-associated membrane protein-associated protein B (vaPB) with OMM protein tyrosine phosphatase-interacting
protein 51 (PtPiP51)254. in cells lacking the vaPB–PtPiP51 complex, autophagy is impaired255,256, in line with the evidence
that MFN2-tethered MerCs are a source of autophagosome membranes257. whether MerC-derived autophagosomes
can engulf any cargo or are specific for one type of autophagy (erphagy, mitophagy, pexophagy, etc.) remains to be
defined. the existence of tethering complexes might suggest that contact sites are stable; however, we can infer that
they can quickly undergo remodelling following tissue-specific metabolic needs or stresses110. MerC plasticity likely
relies on regulatory partners such as the keratin filament-binding protein trichoplein (tCHP; also known as mitostatin),
which interacts with and inhibits MFN2 (ref.258). regulatory components do not necessarily need to be proteins:
ceramides, for example, can bind vDaC2 to regulate apoptosis by blocking vDaC2-mediated retrotranslocation
of the pore-forming proapoptotic protein BaX, thereby allowing its accumulation on mitochondria259.

ER
IP3R MFN2 PTPIP51

Autophagy DISC1 TCHP Lipid and


GRP75

phospholipid
Ca2+ PACS2
biosynthesis
homeostasis
FUNDC1
VAPB Mitochondrial
Apoptosis
morphology
OMM VDAC

Mitochondrion

mitochondria and endosomes carrying iron-bound as shown by enhanced association between RAB5-
transferrin123, indicating that mitochondria function- positive early endosomes and mitochondria during
ally engage in crosstalk with the endosomal system. oxidative stress. This association led to decreased apop-
Cholesterol can also be transferred from endosomes totic potential of mitochondria, likely via protection (for
to mitochondria. The physical coupling of these example, mediated by the transport of lipids and meta­
organelles might depend on the endosomal protein bolites between the organelles) and/or repair of damaged
MLN64: its amino terminus is anchored at the endo- mitochondria to preserve cell viability125. The impact of
somal surface and its carboxy terminus (harbouring a mitochondria and their dynamics on the endosomal
cholesterol-binding domain) faces the cytosol, being system are currently unknown.
Transferrin
An iron-binding glycoprotein
available for OMM interaction and direct cholesterol Lysosomes contribute to mitochondrial homeostasis
that controls the level of free transfer to mitochondria124. These contacts can also be not only by mediating their clearance in stress/unhealthy
iron in the blood. regulated by certain cues, including pathological signals, conditions but also by marking the sites of mitochondrial

www.nature.com/nrm
Reviews

fission95. The lysosomal RAB7 GTPase is fundamental mitochondria-shaping proteins (Table 2) and their links
to keep the two organelles in touch: its displacement by to cellular signalling pathways; reciprocally, we will
RAB7 GTPase-activating protein TBC1D15, which asso- also discuss how these cellular events are influenced by
ciates with FIS1, drives RAB7 inactivation, lowering the changes in mitochondrial morphology.
extent of juxtaposition of mitochondria and lysosomes
and providing space for ER–DRP1-mediated fission Mitochondrial morphology and apoptosis. Historically,
events95. Whether the mitochondria-shaping machin- cell death has been the first cellular process where
ery can locally influence lysosomal functions remains changes in mitochondrial shape have been implicated135.
to be elucidated. However, this observation remained anecdotal until the
discovery of the role of DRP1-dependent mitochondrial
Interactions between lipid droplets and mitochondria. fragmentation2 and of cristae remodelling in cytochrome c
Juxtaposition of mitochondria and lipid droplets has release and apoptosis3 (Fig. 3a,b). We then learned the
been described in adipose and skeletal muscle tissues, basic tenets of how mitochondrial dynamics are con-
and has been shown to be mediated by perilipin 5, which trolled in the course of cell death and how they impact
resides at the surface of lipid droplets126. Mitochondria cell death execution.
associated with lipid droplets (also known as peri­droplet During the initial stages of apoptosis, proapoptotic,
mitochondria) display unique properties, including pore-forming proteins, BAX and BAK, translocate to
increased bioenergetic capacity, lower ß-oxidation and discrete foci on mitochondria, which colocalize with
slower fission–fusion cycles127. Thus, it appears that the DRP1 and MFN2 (but not other mitochondria-shaping
interaction with lipid droplets has an inhibitory effect on proteins) and subsequently become mitochondrial
mitochondrial dynamics, perhaps by directly influenc- fission sites. It was then proposed that these foci are
ing the fusion–fission machinery, or by exposing its core associated with block of fusion, which — by perturb-
players to a steady flux of fatty acids or their derivatives ing the balance between fusion and fission — results in
that might inhibit them, in similarity to MFF inhibition apoptotic mitochondrial fragmentation136.
by ceramide-derived sphingolipids128. Mitochondrial fission is fundamental for apop-
tosis2. BAX and BAK promote DRP1 stabilization at
Interactions between peroxisomes and mitochondria. fission sites, preventing its normal cycling between
Crosstalk between peroxisomes and mitochrodria is the cytosol and mitochondria, and ultimately initiat-
supported by the shared metabolic processes (such as ing apoptotic fragmentation137. This mitochondrial
β-oxidation of fatty acids and bile acid synthesis)129 stabilization is associated with DRP1 sumoylation,
as well as by the shared division machinery components occurring at eight different sites and catalysed by the
(DRP1, MFF and FIS1). Although the molecular details mitochondrial-associated SUMO E3 protein ligase
of this crosstalk remain poorly uncharacterized, both MAPL138,139 (Table 2). During cell death, DRP1 sumoy-
organelles engage in the formation of membrane contact lation and stabilization on mitochondrial membranes
sites. Fzo1p, the yeast mitofusin, serves as a bona fide favours mitochondrial fission and network fragmenta-
tether between peroxisomes and mitochondria130, par- tion, but also contributes to establishment of a func-
alleling the mitochondria-tethering function of mam- tional platform for the transmission of Ca2+ signals from
malian MFN2 with organelles other than mitochondria the ER to mitochondria that support cristae remodel-
such as the ER71 and melanosomes131. A handful of ling, amplify cytochrome c release and fuel mitochon-
other tethers between peroxisomes and mitochondria, drial apoptosis (Fig. 3a; see the next paragraph)137,140.
not related to mitochondrial dynamics, have also been In the context of cell death, mitochondrial fission can
discovered, ranging from peroxisome protein Pex11 and also be regulated by modulating DRP1 Ser637 phospho-
Mdm34, a component of the ER–mitochondria tether- rylation–dephosphorylation. Because Ser637 dephos-
ing complex known as the ER–mitochondria encounter phorylation and DRP1 recruitment to mitochondria
structure (ERMES) in yeast, to ATP-binding cassette depends on calcineurin, which is Ca2+ sensitive, it is not
subfamily D member 1 (ABCD1) and enoyl-CoA delta surprising that several pathological conditions leading
isomerase 2 (ECI2; also known as ACBD2) isoform A in to sustained cytoplasmic Ca2+ influx, such as apop-
mammals132–134. We expect that the discovery of the full tosis, can cause mitochondrial fragmentation 141–147.
set of tethers between mitochondria and peroxisomes Accordingly, calcineurin inhibition can be beneficial
will lead to a deeper understanding of the crosstalk in multiple conditions associated with elevated cell
between mitochondria and their dynamics and perox- death105,148–157.
isomes, providing new insights into the regulation of Inhibition of mitochondrial fusion during apoptosis
cell metabolism. has been linked to the activity of different kinases that
β-Oxidation phosphorylate both mitofusins. MFN1 is phosphoryl-
The catabolic process through
Crosstalk with cell function ated by the mitogen-activated protein kinase cascade
which fatty acids are degraded
in mitochondria to produce As mentioned earlier, the list of cellular processes member extracellular signal-regulated kinase on Thr562
acetyl coenzyme A. impacted by mitochondrial shape changes is long (Table 2). This phosphorylation was shown to directly
and steadily growing (Fig. 1a). We will now provide an inhibit fusion by perturbing MFN1 oligomerization
Ceramide overview of several examples of crucial cellular events and mitochondrial tethering. Non-oligomerized MFN1
A family of lipid molecules
composed of a fatty acid
that are accompanied by mitochondrial dynamics. was then implicated in promoting BAK oligomeriza-
conjugated to the 18-carbon We will discuss how these processes affect mitochon- tion to favour mitochondrial permeabilization dur-
amino alcohol sphingosine. drial morphology, focusing on the many PTMs of ing apoptosis. Inhibition of mitochondrial fusion was

Nature Reviews | Molecular Cell Biology


Reviews

a b Mitochondrial fission
and permeabilization

Apoptotic
Hyperfusion • Increased membrane stimuli
potential
• Ca2+ accumulation

• Altered ultrastructure
• Cytochrome c redistribution
and release Cristae remodelling
Basal
condition
Apoptosis
Fission

Skipping
Starvation Unopposed cell death
fusion

Inhibition of autophagic
degradation
PKA-mediated
phosphorylation

RCC
d Fibroblast iPS cell Differentiated cell DIABLO
OXPHOS Glycolysis OXPHOS Disorganized RCC cAMP
OPA1 Calcium
OPA1 oligomer PKA
Cytochrome c DRP1

Increased Increased APAF1 Phospho-DRP1


fission and fusion
mitophagy Apoptosome Autophagosome
Caspase
Mitophagosome
IAP

Fig. 3 | changes in mitochondrial morphology impact on complex cellular associated Ca2+ influx into mitochondria (Fig. 2). BAX/BAK activation and Ca2+
processes/behaviours. a | Mitochondrial fission is a hallmark of the early influx promote destabilization of optic atrophy protein 1 (OPA1) oligomers at
steps of apoptosis2, and it is associated with altered mitochondrial cristae junctions, resulting in widening of the cristae. This promotes the
ultrastructure (cristae remodelling), leading to intramitochondrial release of cytochrome c. c | Nutrient deprivation (starvation) induces a
cytochrome c redistribution from cristae to the intermembrane space171 response in which mitochondrial fusion plays a key role. Elongation of
and release to the cytosol. Mitochondrial hyperfusion can precede fission mitochondria prevents their degradation by autophagy , thus preserving
during apoptosis2 and can lead to mitochondrial Ca2+ overload212 and metabolic fitness and preventing cell death. This regulation of mitochondrial
mitochondrial ultrastructural damage that promotes cytochrome c release. membrane dynamics occurs via increased cytosolic cAMP levels, which
b | Mitochondrial ultrastructure plays a fundamental role in the control of cell activate protein kinase A (PKA), leading to dynamin-related protein 1 (DRP1)
death. Apoptotic stimuli induce the activation of the proapoptotic Ser637 phosphorylation to inactivate mitochondrial fission. d | Mitochondrial
pore-forming proteins BAX and BAK (not shown), which mediate membrane dynamics regulate cell reprogramming into induced pluripotent
mitochondrial outer membrane permeabilization (MOMP), allowing the stem (iPS) cells as well as stem cell differentiation. Specifically , induction of
release of intramitochondrial metabolites and proteins, prominently pluripotency is associated with mitochondrial fragmentation via regulation
including two caspase activators: cytochrome c and direct inhibitor of of DRP1 phosphorylation and removal of mitochondria via mitophagy266.
apoptosis protein (IAP)-binding protein with low pI (DIABLO; also known as By contrast, early steps of iPS cell differentiation are accompanied by
SMAC). Once in the cytosol, cytochrome c interacts with apoptotic mitochondrial fusion. These changes in mitochondrial membrane dynamics
protease-activating factor 1 (APAF1), which induces the formation of the go hand in hand with metabolic changes: iPS cells are characterized by
apoptosome and subsequent activation of the caspase cascade; this is further reduced oxidative phosphorylation (OXPHOS) and rely mostly on glycolysis,
supported by proteins such as DIABLO, which sequesters caspase inhibitor while in differentiated cells, OXPHOS is a prevailing mechanism providing
proteins. Besides MOMP, apoptosis induces mitochondrial fission and cellular energy. RCC, respiratory chain complex.

www.nature.com/nrm
Reviews

Ischaemia–reperfusion
also observed during heart ischaemia–reperfusion injury All in all, the vast literature supports an essential
injury on phosphorylation of MFN1 by protein kinase C role for mitochondrial membrane dynamics as core
Tissue damage occurring isoform-β2 at Ser86 (Table 2), with detrimental effects mechanisms of cell death. It is thus not surprising that
especially in the heart on on mitochondrial function158. MFN2 is phosphorylated mutations in mitochondria-shaping proteins can cause
coronary artery occlusion and
reperfusion on physicians’
by the stress-induced JUN amino-terminal kinase on various diseases associated with cell loss (Table 1).
intervention, for example, Ser27 (Table 2), ultimately leading to MFN2 ubiquityla-
by percutaneous coronary tion and proteasomal degradation159. Conversely, in the Mitochondrial morphology and mitochondrial qual-
intervention (formerly known event of stress, oxidation of MFN2 at Cys684 — with ity control. A natural ramification of the discovery
as angioplasty with stent).
the formation of intramolecular disulfide bonds resulting that cell death was linked to and controlled by mito-
Omegasomes
in MFN2 oligomerization — stimulates mitochondrial chondrial shape changes was to understand how
Omega-shaped organelles fusion160,161. It would be interesting to understand the mitochondrial membrane dynamics are involved
which precede the formation hierarchic organization of these MFN2 modifications in in mitochondrial quality control mechanisms, which
of autophagosomes. the course of cell death. For example, the massive DRP1- serve as means of adapting the mitochondrial network
dependent mitochondrial fission occurring during to cope with insults. These events range from proteo-
apoptosis is preceded by mitochondrial hyperfusion2 lytic control of mitochondrial protein misfolding, to
(Fig. 3a): perhaps this hyperfusion could be an attempt to transcriptional responses driven by transcription fac-
protect mitochondria against apoptosis and removal via tors that can shuttle between mitochondria and the
autophagy (mitophagy) under stress and could involve nucleus to selective mitochondrial degradation by
oxidative modifications of MFN2. autophagy, also known as mitophagy, and finally to
The second facet of mitochondrial morphological shedding of mitochondrial-derived vesicles (MDVs) or
shape changes during apoptosis is OPA1-dependent mitochondrial-derived compartments (MDCs) (Box 3).
cristae remodelling, whereby the neck of the cristae The interplay between mitochondrial dynamics and
widens, to promote cytochrome c release51,162,163 (Fig. 3b). quality control has been the focus of several excellent
The consensus that cristae remodelling is required for reviews176–180. Here, we will focus our attention on key
cytochrome c release28,49,164–168 was questioned by a report regulatory mechanisms of quality control pathways that
placing cristae remodelling downstream of caspase acti- pertain to mitochondrial membrane dynamics.
vation and hence of cytochrome c release169. However, An important facet of quality control mechanisms
independent studies showed that cristae remodelling is the activation of proteolytic cascades, which function
occurs in cells lacking BAX and BAK, the BCL-2 family to clear the cell of misfolded proteins and to allow their
members essential to allow cytochrome c egress from replacement with newly synthesized polypeptides. One
mitochondria163, as well as in cells where caspases are important pathway of proteolytic degradation involves
inhibited170, further reinforcing that cristae remodelling the ubiquitin–proteasome system, which has emerged as
is required for efficient apoptotic cytochrome c release. a prominent regulator of mitochondrial fusion. The first
The precise mechanism by which cristae remodelling evidence that mitochondrial fusion could be regulated
is induced is still a matter of debate. Proapoptotic mem- came from studies reporting proteasomal degradation of
bers of the BCL-2 family can directly interact with OPA1, the yeast mitofusin orthologue, Fzo1p, which is ubiquity­
causing the disassembly of the oligomers that correlate lated and subsequently targeted to the proteasome by the
with cristae tightness28. In a different mechanism, apop- F-box protein Mdm30p. It was subsequently found that
totic mitochondrial Ca2+ overload (caused among other Drosophila melanogaster mitofusin is the target of the
processes by the stabilization of the ER–mitochondria ubiquitin ligase parkin181 (the encoding gene is mutated
interface) can also lead to cristae remodelling that in autosomal recessive juvenile forms of Parkinson dis-
depends on the permeability transition pore140,171, a ease) which constitutes a crucial component of the mito-
Ca2+-dependent high-conductance IMM channel whose chondrial quality control pathway involving removal of
opening can lead to osmotic swelling of mitochondria mitochondria via mitophagy182,183. In flies, parkin directly
and cristae distension172. Disassembly of OPA1 contain- ubiquitylates Marf (but not Opa1 or Drp1) and its loss
ing oligomers is also a hallmark of this pathway of cristae leads to increased Marf abundance in vivo and concom-
remodelling, but it likely follows the osmotic distension itant elongation of mitochondria181. A similar mecha-
of the IMM. Another possibility is that during cell death, nism operates in mammalian dopaminergic SH-SY5Y
OMA1 protease is hyperactivated — via mitochondrial cells, where parkin ubiquitylates MFN1. It is noteworthy
depolarization or accumulation of reactive oxygen spe- that Parkinson disease-associated mutations impair this
cies (ROS)90,92, or via BAX/BAK activation146 — leading parkin-mediated ubiquitylation of MFN1, which could
to OPA1 cleavage and cristae remodelling. However, explain why mitochondria appear elongated in this cell
whether OPA1 cleavage is sufficient to induce cris- model of Parkinson disease184. Since these original dis-
tae remodelling is unclear. Similarly, it is uncertain if coveries, the field has progressed considerably, and we
cristae remodelling requires also other components of have come to appreciate that several OMM proteins are
the fission machinery, such as MID49/MID51 (ref.170). ubiquitylated and that this is specifically linked to the
In addition to proteolysis, OPA1 is also subject to vari- regulation of mitophagy185. This ubiquitylation is funda-
ous PTMs that include acetylation173, nitrosylation174 and mental for the clearance of defective mitochondria as it
O-linked N-acetylglucosamine glycosylation175 (Table 2) is a triggering signal for the mitophagic process. Other
that have been associated with stress conditions. However, crossroads between mitochondrial dynamics and auto-
it is thus far unclear if these PTMs are responsible phagy exist. For example, mitochondria supply mem-
for OPA1 inactivation and for cristae remodelling. branes for the formation of omegasomes, which occurs at

Nature Reviews | Molecular Cell Biology


Reviews

Box 3 | MDVs and MDcs be potentially modulated by a plethora of additional


non-core proteins to regulate mitophagy. For exam-
the observation of small vesicles excising from mitochondria, named mitochondrial- ple, ganglioside differentiation associated protein 1
derived vesicles (MDvs), was first observed 10 years ago, and since then MDvs have (GDAP1) has been classified as a ‘fission factor’ because
been established as yet another type of interorganelle communication. indeed, MDvs its overexpression leads to DRP1/FIS1-dependent
can fuse with other subcellular compartments, including peroxisomes, endosomes and
mitochondrial fragmentation 189. However, despite
lysosomes, thereby transferring mitochondrial proteins and lipids to these organelles179.
their mitochondrial origin was ascertained by detection of outer mitochondrial these insights, we deem that the space of the regulation
membrane (OMM) proteins, such as mitochondrial-anchored protein ligase (MaPL), of mitochondria-shaping proteins in quality control
inner mitochondrial membrane (iMM) proteins or, in some cases, even mitochondrial mechanisms is truly open for discovery.
matrix molecules, which can be found in the MDv membranes or lumen190,260,261. MDv A recently emerging mechanism of mitochondrial
cargo, however, is not random: it is selectively chosen depending on the mechanism quality control involves the release of MDVs and the
that activates MDv biogenesis. For example, mitochondrial stress induced by reactive generation of MDCs (Box  3). MDVs are small vesi-
oxygen species led to stimulation of MDvs particularly enriched in voltage-dependent cles that originate from mitochondria, engulf certain
anion channel (vDaC; also known as mitochondrial porin), whereas chemical inhibition cargoes and transfer them to other organelles, such as
of respiratory chain complexes induced biogenesis of MDvs carrying the core subunit of lyso­somes and peroxisomes (Fig. 1c). MDVs also influence
respiratory complex iii. Mechanistically, MDvs detach from mitochondria independently
mitochondrial membrane dynamics, from the structural
of dynamin-related protein 1 (DrP1)190,260,262, but require proteins that impinge on
mitochondrial dynamics, such as parkin and PiNK1, for MDvs directed to lysosomes as well as the functional point of view. Structurally, they
and members of the retromer complex, such as vPs35, for MDvs associated with contribute to membrane remodelling by excision of
peroxisomes261. the exact roles of MDvs are not fully understood. However, on the mitochondrial membranes; functionally, they include
one hand, MDvs targeted to peroxisomes may allow transport of specific lipids and/or OMM, IMM and/or matrix proteins that could either be
proteins (including membrane-shaping proteins) from mitochondria. One the other targeted to lysosomes for degradation or translocated to
hand, lysosome-targeted MDvs could represent a means of quality control, whereby peroxisomes (with as of yet unclear consequences)179,190.
damaged mitochondrial portions are removed before they could negatively impact on MDCs appear to be a novel route of mitochondrial qual-
their parental mitochondrion179. Likely, this mechanism would not suffice if the structure ity control, mediating clearance of OMM and/or IMM
and function of that individual organelle are compromised, requiring its entire removal proteins following mitochondrial dysfunction191. While
through mitophagy263.
MDVs and MDCs are certainly involved in mitochon-
apart from MDvs, mitochondria can also bud structures referred to as mitochondrial-
derived compartments (MDCs), which selectively accumulate both OMM and iMM drial quality control, to what extent they definitively
proteins and after release involving dynamin-mediated fission are degraded in the participate in the regulation of mitochondrial dynamics
lysosome. MDCs were described for the first time in yeasts: here, the MDC-mediated remains to be clarified.
degradation pathway is initiated in the presence of vacuole-driven mitochondrial
dysfunctions and could function in mitochondrial quality control, independently of (or Mitochondrial morphology and metabolism. A further
in parallel to) mitophagic clearance. the mechanism of this specific protein degradation field where mitochondrial dynamics are thought to have
via MDCs is not known, but as shown in yeast, it relies on tom70 and tom71 — two a crucial role is the control of metabolism. Elongated
paralogues forming a component of the mitochondrial tOM complex involved in the mitochondria are thought to be more bioenergetically
recognition and initial import steps for all mitochondrially directed proteins191. efficient. This concept perhaps stems from the observa-
although MDCs and MDvs might appear similar, they are not the same entity. First,
tion that mitochondria fragment in the course of apop-
they are likely to engulf different cargos. second, MDCs require the fission machinery for
their release, while MDvs do not. third, they likely have different roles in mitochondrial tosis, as we described earlier, which goes hand in hand
homeostasis, with MDCs primarily acting in selective protein clearance/degradation, with the energetic crisis of cells undergoing cell death.
while MDvs could also mediate cargo exchange and thereby act as signalling entities. In any case, molecular determinants of bioenergetic
Further studies will certainly clarify the mechanisms and physiological functions of MDCs efficiency reside in the IMM and changes in mitochon-
and MDvs as well as their eventual impact on pathological conditions, such as ageing. drial morphology inevitably involve IMM remodelling21.
Indeed, mitochondrial architecture and function were
originally observed in isolated mitochondria undergo-
sites of interaction of mitochondria with the ER, which, ing cycles of respiration and inactivity18. Mitochondria
as described earlier, also serve as sites for mitochondrial are basically thermodynamic converters of energy
fission. Reflecting this association, there is mounting evi- and operate according to the chemiosmotic theory of
dence that parkin ubiquitylates and inhibits the key ER– Mitchell192 (Supplementary Box 1), but this key function
Retromer complex mitochondria tether MFN2 (refs181,186), thus altering the is often forgotten when mitochondrial involvement in
A coat-like complex that crosstalk between these two types of organelles (Box 2) cell biology or in tissue function is being studied. The
mediates the recycling of the and providing a means to modulate both mitophagy and chemiosmotic theory explains and rationalizes very
endosomal cargo into vesicles
moving back to the Golgi
mitochondrial membrane dynamics. It is interesting complex mitochondrial processes such as Ca2+ uptake
apparatus. to note that beyond ubiquitylation, few other PTMs of and extrusion or mitochondrial depolarization during
mitochondria-shaping proteins have been identified apoptotic cytochrome c release. It thus should come as
5′-AMP-activated protein so far in the context of mitochondrial quality control, no surprise that several experimental observations of the
kinase
with the notable exception of MFN2 phosphorylation, consequences of perturbed mitochondrial dynamics can
(AMPK). A key protein kinase
that plays a critical role in which is a prerequisite for MFN2 ubiquitylation and be clarified if one keeps this role as energy converters
cellular homeostasis. By mitophagy187, and the phosphorylation of DRP1 recep- in mind. The link between mitochondrial membrane
phosphorylating a plethora tor MFF by 5′-AMP-activated protein kinase (AMPK), dynamics and mitochondrial metabolism is such an
of targets, it acts as a general which is also a prerequisite for mitochondrial fission and example. The thermodynamic efficiency of mitochondria
switch to regulate cellular
functions such as glucose
elimination by mitophagy188. is fixed, but respiratory chain supercomplexes can stream-
absorption, fat oxidation and Because fission events are required for clearance of line mitochondrial electron transfer and respiration.
mitochondrial biogenesis. damaged mitochondria through mitophagy, fission can Notably, OPA1 overexpression can favour the assembly

www.nature.com/nrm
Reviews

Chemiosmotic theory
and stability of respiratory chain supercomplexes by to mitochondrial fission in early mitosis198. This mitotic
The theory proposed by tightening mitochondrial cristae21. In addition, through fragmentation of the mitochondrial network favours bal-
Mitchell explaining ATP interactions with IMM solute transporters, OPA1 anced mitochondrial redistribution into daughter cells
synthesis by mitochondria. can also act as a sensor of metabolic changes, thereby and ensures their proper function in bioenergetics199.
The chemical energy of NADH
and FADH2 is converted by the
allowing adaptation of cristae shape and respiration to Mitotic mitochondrial fragmentation is also important
electron-transport chain into cellular needs193. to shut down the oxidative mitochondrial metabolism
an electrochemical proton Mitochondrial fission is also regulated to couple in dividing cells; this metabolic rewiring allows glucose
gradient across the inner mitochondrial morphology to the energetic status of the and glutamine-derived metabolic intermediates to be
mitochondrial membrane
cell. This process occurs at the level of DRP1 and of its used for efficient biosynthesis of macromolecules, such
(IMM), whose free energy is
in turn used to catalyse the
main receptor MFF. In conditions of starvation, DRP1 as DNA and lipids, required for cell division200–202.
phosphorylation of ADP by the is phosphorylated on Ser637 in a PKA-dependent fash- By contrast, when cells exit mitosis, DRP1 is ubiqu­
IMM-bound ATP synthase. ion and this leads to unopposed mitochondrial fusion itylated by APC/C (Table 2), which targets DRP1 for
and ultimately in longer mitochondria4,5, which, in turn, proteasomal degradation, thereby shifting mitochon-
Respiratory chain
supercomplexes
prevents their autophagic degradation and supports drial dynamics in interphase cells towards elongation
Assembly of distinct survival of starving cells (Fig. 3c). By contrast, fission is to support normal cell metabolism203,204. In addition,
respiratory chain complexes induced by phosphorylation mediated by AMPK, the as we described earlier when discussing the regulation
that guarantees efficient ATP master kinase that is activated when cellular ATP levels of mitochondrial fission during cell death, DRP1 can
production while reducing
drop. AMPK associates with mitochondria, which may be stabilized on the mitochondrial surface by sumoyl-
electron loss and production
of reactive oxygen species.
allow it to rapidly sense the changes in the energy status ation. This DRP1 sumoylation is antagonistically reg-
of a cell and adapt mitochondrial function (by regulat- ulated by the SUMO protease SENP5 (ref.205), which
Epithelial–mesenchymal ing its biogenesis, quality control and dynamics) to the resides primarily within the nucleoli and during G2/M
transition energy needs194. AMPK directly phosphorylates two sites phase translocates to mitochondria to desumoylate
The process through which
epithelial cells lose their
on MFF, Ser155 and Ser172, which were shown to facili- DRP1 (ref.206). Ultimately, these modifications allow
characteristics of adhesion tate mitochondrial fission downstream of mitochondrial the fine-tuning of mitochondrial morphology during
and polarity and acquire those dysfunction caused by mitochondrial respiration inhi- cell cycle. Specifically, the dynamic regulation of DRP1
typical of mesenchymal stem bition. This mechanism could support the removal of activity by degradation and stabilization on mitochon-
cells, including differentiation
damaged mitochondria via autophagy or induce apop- dria exemplifies how distant cell signalling cascades
abilities
tosis in cells with severely dysfunctional mitochondria188. (cell cycle progression and cell death) can converge on
Nuclear factor of activated Similarly, in the context of ischaemia–reperfusion injury, the same mitochondrial dynamics effector to modu-
T cells which leads to bioenergetic insufficiency by several late mitochondrial shape and ultimately the complex
(NFAT). A family of converging mechanisms195, MFF phosphorylation is decision between life and death.
transcription factors that can
translocate into the nucleus
promoted by serine/threonine kinase casein kinase-2α
in response to stimuli that (CK2α), leading to mitochondrial fission and cell death Mitochondrial morphology and cell differentiation.
enhance intracellular Ca2+ (it is though unclear whether MFF is a direct substrate Differentiation and pluripotency are complex cell
levels. Once in the nucleus, of CK2α)196. responses that previously were not considered to be
they induce transcription of
influenced by mitochondrial membrane dynamics but
target genes.
Mitochondrial morphology and the cell cycle. Mitochon­ were considered rather to be controlled at the level of
drial shape must also be regulated during the cell cycle gene expression. This view changed with the discovery
to allow appropriate partitioning of mitochondria to the that mitochondrial fusion is required for heart devel-
daughter cells. Indeed, while during interphase mito- opment by controlling NOTCH signalling and sub­
chondria appear long and tubular, during early mitosis sequent cardiomyocyte differentiation7. In the following
they are fragmented in a DRP1-dependent fashion197. years the concept of the involvement of dynamic changes
Cell cycle progression depends on a complex interac- in the mitochondrial network in cell fate decisions was
tion among different signal transduction cascades that consolidated and extended.
work as checkpoints for the subsequent mitotic steps Mitochondrial dynamics were observed to deter-
and for cytokinesis. The three most important protein mine the differentiation potential of induced pluri­
families involved in mitosis are the cyclin-dependent potent stem cells. Specifically, the first steps of induced
kinases (CDKs; kinases that bind cyclin-like proteins, pluripotent stem cell differentiation are accompanied
which are responsible for the transition between cell by increased mitochondrial fusion, whereas if fission
cycle stages), the Aurora family (overall controlling prevails, pluripotent stem cells fail to differentiate118,207
chromatid segregation, the formation of mitotic spin- (Fig.  3d) . Mitochondrial hyperfusion (induced by
dle, and centrosome separation) and kinases of the epithelial–mesenchymal transition) appears to correlate
anaphase-promoting complex/cyclosome (APC/C; with increased self-renewal potential in mammary
a multiprotein E3 ubiquitin ligase complex that targets stem cells118. Similarly, mitochondrial fusion orches-
mitotic proteins for degradation, promoting anaphase). trates a NOTCH-dependent self-renewal transcrip-
Notably, cell cycle-dependent changes in the mitochon- tion programme in neural stem cells6. Furthermore,
drial shape are mediated by the activity of members of cell fate decisions in haematopoietic stem cells208 and
all these cell cycle controller families. during osteoclastogenesis209 have been shown to be
In particular, CDK1 (in complex with cyclin B) acts regulated by MFN2, which was essential to control
downstream of Aurora A to phosphorylate DRP1 on transcription activity of nuclear factor of activated T cells
Ser585 and Ser616 (Table 2), which promotes the recruit- (NFAT). Finally, in embryonic stem cells mitochon-
ment of DRP1 to the mitochondrial surface, thus leading drial fusion activation was necessary and sufficient to

Nature Reviews | Molecular Cell Biology


Reviews

Naive embryonic stem cells drive the transition from naive embryonic stem cells to be a source of ROS (Supplementary Box 1), which can
Embryonic stem cells of the primed embryonic stem cells210. Molecularly, in several cause DNA damage, leading to enhancement of ROS
preimplantation blastocyst that cases, Ca2+ appears to link changes in mitochondrial production, DNA damage response and activation of a
have the potential to generate dynamics with altered stem cell differentiation. For transcription programme to induce mitochondrial bio-
all the somatic cell lineages.
example, during cardiomyocyte differentiation, mito- genesis and senescence-associated secretory phenotype213.
Primed embryonic stem chondrial fusion is required to lower entry of Ca2+ from In addition to increased oxidative metabolism, mito-
cells the extracellular space and limit NOTCH signalling7. chondrial hyperfusion would also impede mitophagy,
Embryonic stem cells (from Similarly, in haematopoietic, neuronal and osteoclast eventually leading to accumulation of dysfunctional
post-implantation epiblast) that
progenitors, changes in the shape of mitochondria and/or mitochondria and to increased ROS production215.
can self-renew and differentiate.
their interaction with the ER impact on NOTCH and In line with this, depletion of these organelles can
Senescence-associated NFAT because of sustained cytoplasmic Ca2+ loads6,208,209. reverse most of the phenotypes associated with
secretory phenotype Lastly, in induced pluripotent stem cells, MFF over­ radiation-induced senescence, including enhanced ROS
The release of factors such as expression and increased fission also increase cytosolic production and senescence-associated secretory pheno-
inflammatory cytokines, growth
factors and proteases from
Ca2+ concentrations, leading to hyperactivation of Ca2+/ type. All in all, we think that senescence triggers mito-
senescent cells. calmodulin-dependent kinase II and degradation of chondrial fusion, at least in some specific senescence
the master regulator of stem cell biology β-catenin207. pathways or tissue contexts. However, it is unclear if this
Integrated stress response Notably, mitochondrial dynamics influence Ca2+ sig- process is required for senescence and whether it is of
A generally prosurvival stress
nalling by multiple mechanisms. For example, MFN2 any relevance in vivo. Similarly, it is unknown whether
response induced on exposure
of the cell to either internal
can control ER–mitochondria Ca2+ transfer and hence changes in mitochondrial morphology trigger senes-
factors such as accumulation the amplitude of cytosolic Ca2+ transients, but also the cence only by enhancing ROS production. We expect
of unfolded protein leading to molecular machinery that allows refilling of the intra- that the connection between mitochondrial morphology
endoplasmic reticulum stress cellular Ca2+ stores71,119. By contrast, in the absence of and senescence will be extensively explored in the future,
or external factors such as
nutrient deprivation and viral
mitochondrial fission, mitochondria take up excess Ca2+ especially given the interest in senescence-modulating
infection. (refs211,212), thereby lowering cytoplasmic Ca2+ transients. drugs in ageing and cancer.
Thus, it is impossible to conclude that, for example,
ATF4 mitochondrial fission promotes increased cytoplasmic Roles in organismal homeostasis
Cyclic AMP-dependent
Ca2+ levels whereas mitochondrial fusion lowers them, The availability of molecular tools to control mitochon-
transcription factor 4 plays
a key role in cell response to
and the link between dynamics and Ca2+ transients must drial shape allowed us to understand that the impact
stress stimuli, mostly by be addressed on a case-by-case basis. of the form–function relationship of these organelles
promoting cell survival. It is also interesting to note that overall, these studies extends over the cellular boundaries — changes in the
report effects of mitochondrial shape on differentiation physiology of a certain cell type modifies its crosstalk
Mitokine
A cytokine produced by tissues
or pluripotency but do not address how changes in mito- with surrounding cells, thus influencing the whole organ
undergoing mitochondrial chondrial dynamics are embedded in these complex cas- and even organism physiology and, in some cases, lead-
stress and affecting other cell cades. In other words, whether key factors that regulate ing to pathological conditions or ageing. For example,
types in an endocrine or a differentiation and pluripotency operate by means other the drop in muscle OPA1 levels observed in seden-
paracrine manner. Mitokines
than transcriptional regulation of mitochondria-shaping tary elderly individuals can be modelled in mice with
include, for example, FGF21
and GDF15.
proteins208 is currently unknown. This is perhaps due to inducible OPA1 muscle deletion. In this model, lack of
the very nascent nature of research in this field and to the muscle OPA1 leads to systemic metabolism changes
Leptin current mostly correlative links between mitochondrial and to senescence of distant epithelial tissues by acti-
This term derives from Greek shape and stem cell fate regulation. vating an integrated stress response and ATF4-dependent
leptos (‘thin’); it refers to a
hormone that inhibits hunger.
muscle production and secretion of fibroblast growth
Mitochondrial morphology and cell senescence. A very factor 21 (FGF21), which functions as a mitokine216–218.
Pro-opiomelanocortin recent development in the cellular biology of mito- Similarly, abnormal mitochondrial elongation caused by
(POMC) neurons chondrial dynamics is their key, yet only partially artificial or pathological DRP1 ablation in hippocampal
Neurons located in the
elucidated, role in senescence. Although senescence neurons led to increased ATF4 expression levels and
arcuate nucleus within
the hypothalamus; they
can be broadly defined as the state in which cell pro- finally neuronal FGF21 synthesis219. The discovery that
produce a polypeptide liferation is irreversibly arrested, it should not be con- mitochondrial shape is embedded in systemic signalling
named pro-opiomelanocortin, sidered as a uniform process; rather, different types of was surprising, because the observed transcriptional
whose proteolysis generates tissue-specific senescence pathways exist. In addition modifications and systemic signals occurred before any
several peptide hormones
(including α-melanocyte-
to the cell intrinsic (replicative) senescence, charac- detectable mitochondrial dysfunction216, a finding reca-
stimulating hormone and terized by telomere shortening due to reiterated rep- pitulated in other settings where FGF21 was produced
adrenocorticotropic hormone). licative cycles and chronic DNA damage response, before mitochondrial function was affected220.
senescence can also be caused by oxidative stress, radia- Another relevant example of how mitochondrial
ER stress
tion or oncogenic factors. Changes in the shape and/or dynamics can control homeostatic functions is the
A set of intracellular pathways
activated by the cell to cope
mass of mitochondria appear to underlie both types connection between MFN2, leptin resistance and
with abnormal accumulation of senescence. In human endothelial cells undergoing obesity. Specific ablation of MFN2 in hypothalamic
of unfolded proteins in the intrinsic or radiation-induced senescence, mitochondria pro-opiomelanocortin (POMC) neurons that respond to
lumen of the endoplasmic appear extremely elongated, due to downregulation of leptin to control appetite impaired both the morpho­
reticulum (ER).
DRP1 and FIS1 (refs213,214). In this context, as well as logy of mitochondria and their interactions with the ER.
in oncogene-mediated senescence213, mitochondrial This elicited ER stress, resulting in leptin resistance, and
OXPHOS seems to be a key player in senescence induc- finally severe obesity221. The cellular and systemic meta­
tion. This is likely because mitochondrial respiration can bolic control exerted by MFN2 has also been reported

www.nature.com/nrm
Reviews

Brown adipocytes
in brown adipocytes. Specific ablation of MFN2 in these and that disturbed dynamics of mitochondrial mem-
Cells that compose the brown cells decreased not only mitochondrial length but also branes are linked to several common diseases (Table 1).
adipose tissue, enriched in the extent of the interaction of mitochondria with lipid Nevertheless, despite intense research in this exciting
mitochondria and droplets and cell size. The higher adiposity and body area, several questions remain open. For example, a
characterized by high
weight that characterizes this experimental model deeper understanding of the mechanisms and regulation
thermogenic potential.
can be explained by considering that reduced contact of molecules involved in fusion and fission is required.
Alveolar type 2 epithelial between the two organelles likely reduces the transfer This will call for identification of new players and char-
cells of fatty acids from lipid droplets to mitochondria, and acterization of the structure of already identified ones,
One of the two cell types that
hence halts their utilization as an energy source dur- as well as of PTMs and the dynamics of large protein
build the alveolar epithelium,
responsible for surfactant
ing β-oxidation in favour of their storage, even with a complexes involved in mitochondrial morphology.
synthesis and secretion. normal diet222. One important avenue for future research is to under-
The fission–fusion balance appears fundamen- stand how these mechanisms adapt to different meta-
tal also for the physiology of lungs and muscle. In the bolic and signalling states of the cell. Similarly, the field
first case, mitochondrial fusion is required in alveolar awaits screens to comprehensively map structural teth-
type 2 epithelial cells for production of surfactant, a lipo- ers and regulators of the interfaces between mitochon-
protein complex composed of several phospholipids and dria and other organelles, as well as discovery screens to
cholesterol necessary to reduce surface tension within identify other essential proteins of the fusion and fission
alveoli and preserve their structure. Ablation of MFN1 machinery. Last, but not least, identification of small
or MFN2 indeed impaired alveolar phospholipid syn- molecules able to control mitochondrial dynamics/
thesis and caused spontaneous onset of lung fibrosis223. shape through chemical compound screenings and bio-
In the skeletal muscle, mitochondrial fission determines chemistry approaches is currently being undertaken by
myofibre mass by regulating their survival: mice sub- several laboratories224–226.
jected to transient (duration of 50 days) DRP1 ablation We expect that the information ensuing from these
were characterized by muscle atrophy and functional studies will be used to expand our understanding of the
decline, which was associated with changes to mito- relationship between mitochondrial shape and func-
chondrial morphology (bigger but fewer mitochondria), tion, including its roles in regulating cell metabolism,
functional decline of mitochondria and mitochondrial signalling networks (such as those involved in cell death,
Ca2+ overload, leading to myofibre death211. immunity and stress responses) and even gene expres-
sion through mitochondria–nuclear communication227.
Conclusions and perspectives It will also open up avenues for therapeutic modulation
The field of mitochondrial membrane dynamics has of mitochondrial dynamics in pathological conditions,
expanded tremendously since its inception less than including the associated congenital disorders224–226, dis-
15 years ago. We think that this is mostly due to the eases linked to cell death such as neurodegeneration and
realization that mitochondrial membrane dynamics are stroke224–226 and also cancer228.
embedded in cell signalling networks and has impor-
tant cellular functions (shape–function relationship) Published online xx xx xxxx

1. Liu, X., Kim, C. N., Yang, J., Jemmerson, R. & Wang, X. 10. Collins, T. J., Berridge, M. J., Lipp, P. & Bootman, M. D. with change in metabolic steady state in isolated liver
Induction of apoptotic program in cell-free extracts: Mitochondria are morphologically and functionally mitochondria. J. Cell Biol. 30, 269–297 (1966).
requirement for dATP and cytochrome c. Cell 86, heterogeneous within cells. EMBO J. 21, 1616–1627 A classic article showing the link between
147–157 (1996). (2002). mitochondrial morphological changes and function.
2. Frank, S. et al. The role of dynamin-related protein 1, 11. Kasahara, A. & Scorrano, L. Mitochondria: from cell 19. Mannella, C. A. et al. Topology of the mitochondrial
a mediator of mitochondrial fission, in apoptosis. death executioners to regulators of cell differentiation. inner membrane: dynamics and bioenergetic
Dev. Cell 1, 515–525 (2001). Trends Cell Biol. 24, 761–770 (2014). implications. IUBMB Life 52, 93–100 (2001).
3. Scorrano, L. et al. A distinct pathway remodels 12. Pernas, L. & Scorrano, L. Mito-morphosis: 20. Hackenbrock, C. R., Schneider, H., Lemasters, J. J.
mitochondrial cristae and mobilizes cytochrome c mitochondrial fusion, fission, and cristae remodeling & Hochli, M. Relationships between bilayer lipid,
during apoptosis. Dev. Cell 2, 55–67 (2002). as key mediators of cellular function. Annu. Rev. motional freedom of oxidoreductase components, and
References 2 and 3 provide the first demonstration Physiol. 78, 505–531 (2016). electron transfer in the mitochondrial inner membrane.
that mitochondrial dynamics are essential for 13. Frey, T. G. & Mannella, C. A. The internal structure Adv. Exp. Med. Biol. 132, 245–263 (1980).
apoptosis. of mitochondria. Trends Biochem. Sci. 25, 319–324 21. Cogliati, S. et al. Mitochondrial cristae shape
4. Gomes, L. C., Di, B. G. & Scorrano, L. During (2000). determines respiratory chain supercomplexes
autophagy mitochondria elongate, are spared from 14. Vogel, F., Bornhovd, C., Neupert, W. & Reichert, A. S. assembly and respiratory efficiency. Cell 155,
degradation and sustain cell viability. Nat. Cell Biol. Dynamic subcompartmentalization of the 160–171 (2013).
13, 589–598 (2011). mitochondrial inner membrane. J. Cell Biol. 175, 22. Chandel, N. S. Evolution of mitochondria as signaling
5. Rambold, A. S., Kostelecky, B., Elia, N. & 237–247 (2006). organelles. Cell Metab. 22, 204–206 (2015).
Lippincott-Schwartz, J. Tubular network formation 15. Gilkerson, R. W., Selker, J. M. & Capaldi, R. A. 23. Bahat, A. & Gross, A. Mitochondrial plasticity in cell
protects mitochondria from autophagosomal The cristal membrane of mitochondria is the principal fate regulation. J. Biol. Chem. 294, 13852–13863
degradation during nutrient starvation. Proc. Natl site of oxidative phosphorylation. FEBS Lett. 546, (2019).
Acad. Sci. USA 108, 10190–10195 (2011). 355–358 (2003). 24. Nguyen, M., Millar, D. G., Yong, V. W., Korsmeyer, S. J.
6. Khacho, M. et al. Mitochondrial dynamics impacts 16. Demongeot, J., Glade, N., Hansen, O. & Moreira, A. & Shore, G. C. Targeting of Bcl-2 to the mitochondrial
stem cell identity and fate decisions by regulating a An open issue: the inner mitochondrial membrane outer membrane by a COOH-terminal signal anchor
nuclear transcriptional program. Cell Stem Cell 19, (IMM) as a free boundary problem. Biochimie 89, sequence. J. Biol. Chem. 268, 25265–25268 (1993).
232–247 (2016). 1049–1057 (2007). 25. Scorrano, L. & Korsmeyer, S. J. Mechanisms of
7. Kasahara, A., Cipolat, S., Chen, Y., Dorn, G. W. 17. Wolf, D. M. et al. Individual cristae within the same cytochrome c release by proapoptotic BCL-2 family
& Scorrano, L. Mitochondrial fusion directs mitochondrion display different membrane potentials members. Biochem. Biophys. Res. Commun. 304,
cardiomyocyte differentiation via calcineurin and and are functionally independent. EMBO J. 38, 437–444 (2003).
Notch signaling. Science 342, 734–737 (2013). e101056 (2019). 26. Karbowski, M., Norris, K. L., Cleland, M. M., Jeong, S. Y.
8. Yasukawa, K. et al. Mitofusin 2 inhibits mitochondrial Demonstration that individual cristae can & Youle, R. J. Role of Bax and Bak in mitochondrial
antiviral signaling. Sci. Signal. 2, ra47 (2009). be regarded as independent mitochondrial morphogenesis. Nature 443, 658–662 (2006).
9. Benda, C. Ueber die Spermatogenese der Vertebraten subcompartments. 27. Karbowski, M. et al. Spatial and temporal association
und höherer Evertebraten, II. Theil: die Histiogenese 18. Hackenbrock, C. R. Ultrastructural bases for of Bax with mitochondrial fission sites, Drp1, and
der Spermien. Arch. Anat. Physiol. 73, 393–398 metabolically linked mechanical activity in Mfn2 during apoptosis. J. Cell Biol. 159, 931–938
(1898). mitochondria. I. reversible ultrastructural changes (2002).

Nature Reviews | Molecular Cell Biology


Reviews

28. Landes, T. et al. The BH3-only Bnip3 binds to the supracomplexes. J. Biol. Chem. 281, 13990–13998 OPA1 and cardiolipin. Nat. Cell Biol. 19, 856–863
dynamin Opa1 to promote mitochondrial (2006). (2017).
fragmentation and apoptosis by distinct mechanisms. 51. Frezza, C. et al. OPA1 controls apoptotic cristae 75. Herlan, M., Vogel, F., Bornhovd, C., Neupert, W.
EMBO Rep. 11, 459–465 (2010). remodeling independently from mitochondrial fusion. & Reichert, A. S. Processing of Mgm1 by the
29. Lefkimmiatis, K., Leronni, D. & Hofer, A. M. The inner Cell 126, 177–189 (2006). rhomboid-type protease Pcp1 is required for
and outer compartments of mitochondria are sites of 52. Griparic, L., van der Wel, N. N., Orozco, I. J., maintenance of mitochondrial morphology and of
distinct cAMP/PKA signaling dynamics. J. Cell Biol. Peters, P. J. & van der Bliek, A. M. Loss of the mitochondrial DNA. J. Biol. Chem. 278, 27781–27788
202, 453–462 (2013). intermembrane space protein Mgm1/OPA1 induces (2003).
30. Burdyga, A. et al. Phosphatases control swelling and localized constrictions along the lengths 76. McQuibban, G. A., Saurya, S. & Freeman, M.
PKA-dependent functional microdomains at the outer of mitochondria. J. Biol. Chem. 279, 18792–18798 Mitochondrial membrane remodelling regulated by a
mitochondrial membrane. Proc. Natl Acad. Sci. USA (2004). conserved rhomboid protease. Nature 423, 537–541
115, E6497–E6506 (2018). 53. Glytsou, C. et al. Optic atrophy 1 is epistatic to the (2003).
31. Means, C. K. et al. An entirely specific type I A-kinase core MICOS component MIC60 in mitochondrial 77. DeVay, R. M. et al. Coassembly of Mgm1 isoforms
anchoring protein that can sequester two molecules cristae shape control. Cell Rep. 17, 3024–3034 requires cardiolipin and mediates mitochondrial inner
of protein kinase A at mitochondria. Proc. Natl Acad. (2016). membrane fusion. J. Cell Biol. 186, 793–803 (2009).
Sci. USA 108, E1227–E1235 (2011). 54. Quintana-Cabrera, R. et al. The cristae modulator 78. Griparic, L., Kanazawa, T. & van der Bliek, A. M.
32. Castanier, C., Garcin, D., Vazquez, A. & Arnoult, D. optic atrophy 1 requires mitochondrial ATP synthase Regulation of the mitochondrial dynamin-like protein
Mitochondrial dynamics regulate the RIG-I-like receptor oligomers to safeguard mitochondrial function. Opa1 by proteolytic cleavage. J. Cell Biol. 178,
antiviral pathway. EMBO Rep. 11, 133–138 (2010). Nat. Commun. 9, 3399 (2018). 757–764 (2007).
33. Graham, T. R. & Kozlov, M. M. Interplay of proteins 55. Faelber, K. et al. Structure and assembly of the 79. Baker, M. J. et al. Stress-induced OMA1 activation
and lipids in generating membrane curvature. mitochondrial membrane remodelling GTPase Mgm1. and autocatalytic turnover regulate OPA1-dependent
Curr. Opin. Cell Biol. 22, 430–436 (2010). Nature 571, 429–433 (2019). mitochondrial dynamics. EMBO J. 33, 578–593
34. Friedman, J. R., Mourier, A., Yamada, J., 56. Meeusen, S. et al. Mitochondrial inner-membrane (2014).
McCaffery, J. M. & Nunnari, J. MICOS coordinates fusion and crista maintenance requires the dynamin- 80. Ehses, S. et al. Regulation of OPA1 processing and
with respiratory complexes and lipids to establish related GTPase Mgm1. Cell 127, 383–395 (2006). mitochondrial fusion by m-AAA protease isoenzymes
mitochondrial inner membrane architecture. eLife 4, 57. Amutha, B., Gordon, D. M., Gu, Y. & Pain, D. A novel and OMA1. J. Cell Biol. 187, 1023–1036 (2009).
e07739 (2015). role of Mgm1p, a dynamin-related GTPase, in ATP 81. Head, B., Griparic, L., Amiri, M., Gandre-Babbe, S. &
35. Guarani, V. et al. QIL1 is a novel mitochondrial protein synthase assembly and cristae formation/ van der Bliek, A. M. Inducible proteolytic inactivation
required for MICOS complex stability and cristae maintenance. Biochem. J. 381, 19–23 (2004). of OPA1 mediated by the OMA1 protease in
morphology. eLife 4, e06265 (2015). 58. Sesaki, H., Southard, S. M., Yaffe, M. P. & Jensen, R. E. mammalian cells. J. Cell Biol. 187, 959–966 (2009).
36. Huynen, M. A., Muhlmeister, M., Gotthardt, K., Mgm1p, a dynamin-related GTPase, is essential 82. Cipolat, S. et al. Mitochondrial rhomboid PARL
Guerrero-Castillo, S. & Brandt, U. Evolution and for fusion of the mitochondrial outer membrane. regulates cytochrome c release during apoptosis via
structural organization of the mitochondrial contact Mol. Biol. Cell 14, 2342–2356 (2003). OPA1-dependent cristae remodeling. Cell 126,
site (MICOS) complex and the mitochondrial 59. Wang, L. et al. FAM92A1 is a BAR domain protein 163–175 (2006).
intermembrane space bridging (MIB) complex. required for mitochondrial ultrastructure and function. 83. Saita, S. et al. PARL mediates Smac proteolytic
Biochim. Biophys. Acta 1863, 91–101 (2015). J. Cell Biol. 218, 97–111 (2019). maturation in mitochondria to promote apoptosis.
37. Barbot, M. et al. Mic10 oligomerizes to bend 60. Chen, H. et al. Titration of mitochondrial fusion Nat. Cell Biol. 19, 318–328 (2017).
mitochondrial inner membranes at cristae junctions. rescues Mff-deficient cardiomyopathy. J. Cell Biol. 84. Botham, A. et al. Global interactome mapping of
Cell Metab. 21, 756–763 (2015). 211, 795–805 (2015). mitochondrial intermembrane space proteases
38. Bohnert, M. et al. Central role of Mic10 in the 61. Labbe, K., Murley, A. & Nunnari, J. Determinants and identifies a novel function for HTRA2. Proteomics
mitochondrial contact site and cristae organizing functions of mitochondrial behavior. Annu. Rev. Cell 19, 1900139 (2019).
system. Cell Metab. 21, 747–755 (2015). Dev. Biol. 30, 357–391 (2014). 85. Smirnova, E., Griparic, L., Shurland, D. L. & van der
39. Rabl, R. et al. Formation of cristae and crista junctions 62. Friedman, J. R. & Nunnari, J. Mitochondrial form and Bliek, A. M. Dynamin-related protein Drp1 is required
in mitochondria depends on antagonism between Fcj1 function. Nature 505, 335–343 (2014). for mitochondrial division in mammalian cells.
and Su e/g. J. Cell Biol. 185, 1047–1063 (2009). 63. Tilokani, L., Nagashima, S., Paupe, V. & Prudent, J. Mol. Biol. Cell 12, 2245–2256 (2001).
Identification of the role of MIC60 in cristae Mitochondrial dynamics: overview of molecular 86. Otera, H. et al. Mff is an essential factor for
biogenesis. mechanisms. Essays Biochem. 62, 341–360 (2018). mitochondrial recruitment of Drp1 during
40. Zerbes, R. M. et al. Role of MINOS in mitochondrial 64. Zuchner, S. et al. Mutations in the mitochondrial mitochondrial fission in mammalian cells. J. Cell Biol.
membrane architecture: cristae morphology and GTPase mitofusin 2 cause Charcot-Marie-Tooth 191, 1141–1158 (2010).
outer membrane interactions differentially depend neuropathy type 2A. Nat. Genet. 36, 449–451 87. James, D. I., Parone, P. A., Mattenberger, Y. &
on mitofilin domains. J. Mol. Biol. 422, 183–191 (2004). Martinou, J. C. hFis1, a novel component of the
(2012). 65. Santel, A. & Fuller, M. T. Control of mitochondrial mammalian mitochondrial fission machinery. J. Biol.
41. John, G. B. et al. The mitochondrial inner membrane morphology by a human mitofusin. J. Cell Sci. 114, Chem. 278, 36373–36379 (2003).
protein mitofilin controls cristae morphology. 867–874 (2001). 88. Palmer, C. S. et al. MiD49 and MiD51, new
Mol. Biol. Cell 16, 1543–1554 (2005). 66. Alexander, C. et al. OPA1, encoding a dynamin-related components of the mitochondrial fission machinery.
42. Hessenberger, M. et al. Regulated membrane GTPase, is mutated in autosomal dominant optic EMBO Rep. 12, 565–573 (2011).
remodeling by Mic60 controls formation of atrophy linked to chromosome 3q28. Nat. Genet. 26, 89. Loson, O. C., Song, Z., Chen, H. & Chan, D. C. Fis1,
mitochondrial crista junctions. Nat. Commun. 8, 211–215 (2000). Mff, MiD49, and MiD51 mediate Drp1 recruitment
15258 (2017). 67. Delettre, C. et al. Nuclear gene OPA1, encoding a in mitochondrial fission. Mol. Biol. Cell 24, 659–667
43. Tarasenko, D. et al. The MICOS component Mic60 mitochondrial dynamin-related protein, is mutated (2013).
displays a conserved membrane-bending activity that in dominant optic atrophy. Nat. Genet. 26, 207–210 90. Kalia, R. et al. Structural basis of mitochondrial
is necessary for normal cristae morphology. J. Cell (2000). receptor binding and constriction by DRP1. Nature
Biol. 216, 889–899 (2017). References 66 and 67 provide the first 558, 401–405 (2018).
44. Eydt, K., Davies, K. M., Behrendt, C., Wittig, I. & demonstration of a genetic disorder caused 91. Griffin, E. E., Graumann, J. & Chan, D. C. The WD40
Reichert, A. S. Cristae architecture is determined by by mutant mitochondria-shaping proteins. protein Caf4p is a component of the mitochondrial
an interplay of the MICOS complex and the F1FO ATP 68. Cipolat, S. O., Martins de Brito, O., Dal Zilio, B. & fission machinery and recruits Dnm1p to
synthase via Mic27 and Mic10. Microb. Cell 4, Scorrano, L. OPA1 requires mitofusin 1 to promote mitochondria. J. Cell Biol. 170, 237–248 (2005).
259–272 (2017). mitochondrial fusion. Proc. Natl Acad. Sci. USA 101, 92. Karren, M. A., Coonrod, E. M., Anderson, T. K. &
45. Darshi, M. et al. ChChd3, an inner mitochondrial 15927–15932 (2004). Shaw, J. M. The role of Fis1p-Mdv1p interactions in
membrane protein, is essential for maintaining crista 69. Rojo, M., Legros, F., Chateau, D. & Lombes, A. mitochondrial fission complex assembly. J. Cell Biol.
integrity and mitochondrial function. J. Biol. Chem. Membrane topology and mitochondrial targeting 171, 291–301 (2005).
286, 2918–2932 (2011). of mitofusins, ubiquitous mammalian homologs of 93. Naylor, K. et al. MDV1 interacts with assembled
46. Davies, K. M., Anselmi, C., Wittig, I., Faraldo-Gomez, the transmembrane GTPase Fzo. J. Cell Sci. 115, DNM1 to promote mitochondrial division. J. Biol.
J. D. & Kuhlbrandt, W. Structure of the yeast 1663–1674 (2002). Chem. 281, 2177–2183 (2006).
F1Fo-ATP synthase dimer and its role in shaping the 70. Ishihara, N., Eura, Y. & Mihara, K. Mitofusin 1 and 2 94. Alirol, E. et al. The mitochondrial fission protein hFis1
mitochondrial cristae. Proc. Natl Acad. Sci. USA 109, play distinct roles in mitochondrial fusion reactions requires the endoplasmic reticulum gateway to induce
13602–13607 (2012). via GTPase activity. J. Cell Sci. 117, 6535–6546 apoptosis. Mol. Biol. Cell 17, 4593–4605 (2006).
47. Strauss, M., Hofhaus, G., Schroder, R. R. & (2004). 95. Wong, Y. C., Ysselstein, D. & Krainc, D. Mitochondria–
Kuhlbrandt, W. Dimer ribbons of ATP synthase shape 71. de Brito, O. M. & Scorrano, L. Mitofusin 2 tethers lysosome contacts regulate mitochondrial fission via
the inner mitochondrial membrane. EMBO J. 27, endoplasmic reticulum to mitochondria. Nature 456, RAB7 GTP hydrolysis. Nature 554, 382 (2018).
1154–1160 (2008). 605–610 (2008). 96. Yu, R., Jin, S.-B., Lendahl, U., Nistér, M. & Zhao, J.
48. Paumard, P. et al. The ATP synthase is involved in Identification of the first structural ER–mitochondria Human Fis1 regulates mitochondrial dynamics
generating mitochondrial cristae morphology. tether in mammals. through inhibition of the fusion machinery. EMBO J.
EMBO J. 21, 221–230 (2002). 72. Qi, Y. et al. Structures of human mitofusin 1 provide 38, e99748 (2019).
First demonstration of a role for ATP synthase insight into mitochondrial tethering. J. Cell Biol. 215, 97. Ishihara, N. et al. Mitochondrial fission factor Drp1
in mitochondrial cristae morphology. 621–629 (2016). is essential for embryonic development and synapse
49. Campanella, M. et al. Regulation of mitochondrial 73. Brandt, T., Cavellini, L., Kühlbrandt, W. & Cohen, M. formation in mice. Nat. Cell Biol. 11, 958–966
structure and function by the F1Fo-ATPase inhibitor M. A mitofusin-dependent docking ring complex (2009).
protein, IF1. Cell Metab. 8, 13–25 (2008). triggers mitochondrial fusion in vitro. eLife 5, e14618 98. Osellame, L. D. et al. Cooperative and independent
50. Bornhovd, C., Vogel, F., Neupert, W. & Reichert, A. S. (2016). roles of the Drp1 adaptors Mff, MiD49 and MiD51
Mitochondrial membrane potential is dependent on 74. Ban, T. et al. Molecular basis of selective in mitochondrial fission. J. Cell Sci. 129, 2170–2181
the oligomeric state of F1F0-ATP synthase mitochondrial fusion by heterotypic action between (2016).

www.nature.com/nrm
Reviews

99. Bohuszewicz, O. & Low, H. H. Structure of a binding specificities. J. Cell Biol. 213, 513–524 145. Cho, D. H. et al. S-nitrosylation of Drp1 mediates
mitochondrial fission dynamin in the closed (2016). beta-amyloid-related mitochondrial fission and
conformation. Nat. Struct. Mol. Biol. 25, 722–731 121. Steinman, R. M., Mellman, I. S., Muller, W. A. & neuronal injury. Science 324, 102–105 (2009).
(2018). Cohn, Z. A. Endocytosis and the recycling of plasma 146. Liot, G. et al. Complex II inhibition by 3-NP causes
100. Lee, J. E., Westrate, L. M., Wu, H., Page, C. & membrane. J. Cell Biol. 96, 1–27 (1983). mitochondrial fragmentation and neuronal cell death.
Voeltz, G. K. Multiple dynamin family members 122. Huotari, J. & Helenius, A. Endosome maturation. Cell Death Differ. 16, 899–909 (2009).
collaborate to drive mitochondrial division. Nature EMBO J. 30, 3481–3500 (2011). 147. Brooks, C., Wei, Q., Cho, S. G. & Dong, Z. Regulation
540, 139–143 (2016). 123. Das, A., Nag, S., Mason, A. B. & Barroso, M. M. of mitochondrial dynamics in acute kidney injury in cell
101. Fonseca, T. B., Sánchez-Guerrero, Á., Milosevic, I. Endosome-mitochondria interactions are modulated culture and rodent models. J. Clin. Invest 119,
& Raimundo, N. Mitochondrial fission requires DRP1 by iron release from transferrin. J. Cell Biol. 214, 1275–1285 (2009).
but not dynamins. Nature 570, E34–E42 (2019). 831–845 (2016). 148. Zhang, Z., Liu, L., Wu, S. & Xing, D. Drp1, Mff, Fis1,
102. Kamerkar, S. C., Kraus, F., Sharpe, A. J., Pucadyil, T. J. 124. Charman, M., Kennedy, B. E., Osborne, N. & and MiD51 are coordinated to mediate mitochondrial
& Ryan, M. T. Dynamin-related protein 1 has Karten, B. MLN64 mediates egress of cholesterol fission during UV irradiation–induced apoptosis.
membrane constricting and severing abilities from endosomes to mitochondria in the absence of FASEB J. 30, 466–476 (2016).
sufficient for mitochondrial and peroxisomal fission. functional Niemann-Pick type C1 protein. J. Lipid Res. 149. Li, G. et al. Mitochondrial translocation and
Nat. Commun. 9, 5239 (2018). 51, 1023–1034 (2010). interaction of cofilin and Drp1 are required for
103. Cribbs, J. T. & Strack, S. Reversible phosphorylation 125. Hsu, F. et al. Rab5 and Alsin regulate stress-activated erucin-induced mitochondrial fission and apoptosis.
of Drp1 by cyclic AMP-dependent protein kinase and cytoprotective signaling on mitochondria. eLife 7, Oncotarget 6, 1834–1849 (2015).
calcineurin regulates mitochondrial fission and cell e32282 (2018). 150. Chou, C. H. et al. GSK3beta-mediated Drp1
death. EMBO Rep. 8, 939–944 (2007). 126. Wang, H. et al. Perilipin 5, a lipid droplet-associated phosphorylation induced elongated mitochondrial
104. Chang, C. R. & Blackstone, C. Cyclic AMP-dependent protein, provides physical and metabolic linkage to morphology against oxidative stress. PLoS One 7,
protein kinase phosphorylation of Drp1 regulates its mitochondria. J. Lipid Res. 52, 2159–2168 (2011). e49112 (2012).
GTPase activity and mitochondrial morphology. J. Biol. 127. Benador, I. Y. et al. Mitochondria bound to lipid 151. Torres, G. et al. Glucagon-like peptide-1 inhibits
Chem. 282, 21583–21587 (2007). droplets have unique bioenergetics, composition, vascular smooth muscle cell dedifferentiation through
References 103 and 104 provide the first evidence and dynamics that support lipid droplet expansion. mitochondrial dynamics regulation. Biochem.
for a functional PTM in a mitochondria-shaping Cell Metab. 27, 869–885.e6 (2018). Pharmacol. 104, 52–61 (2016).
protein. 128. Hammerschmidt, P. et al. CerS6-derived sphingolipids 152. Li, A. et al. Metformin and resveratrol inhibit Drp1-
105. Cereghetti, G. M. et al. Dephosphorylation by interact with Mff and promote mitochondrial mediated mitochondrial fission and prevent ER
calcineurin regulates translocation of Drp1 to fragmentation in obesity. Cell 177, 1536–1552.e23 stress-associated NLRP3 inflammasome activation in
mitochondria. Proc. Natl Acad. Sci. USA 105, (2019). the adipose tissue of diabetic mice. Mol. Cell. Endocrinol.
15803–15808 (2008). 129. Fransen, M., Lismont, C. & Walton, P. The peroxisome- 434, 36–47 (2016).
106. Yu, R. et al. The phosphorylation status of Ser-637 in mitochondria connection: how and why? Int. J. Mol. Sci. 153. Lavie, J. et al. Mitochondrial morphology and cellular
dynamin-related protein 1 (Drp1) does not determine 18, E1126 (2017). distribution are altered in SPG31 patients and are
Drp1 recruitment to mitochondria. J. Biol. Chem. 294, 130. Shai, N. et al. Systematic mapping of contact sites reveals linked to DRP1 hyperphosphorylation. Hum. Mol.
17262–17277 (2019). tethers and a function for the peroxisome-mitochondria Genet. 26, 674–685 (2016).
107. Friedman, J. R. et al. ER tubules mark sites of contact. Nat. Commun. 9, 1761 (2018). 154. Li, J. et al. Pharmacological activation of AMPK
mitochondrial division. Science 334, 358–362 131. Daniele, T. et al. Mitochondria and melanosomes prevents Drp1-mediated mitochondrial fission and
(2011). establish physical contacts modulated by Mfn2 alleviates endoplasmic reticulum stress-associated
Identification of the association between ER and and involved in organelle biogenesis. Curr. Biol. 24, endothelial dysfunction. J. Mol. Cell. Cardiol. 86,
mitochondrial fission sites. 393–403 (2014). 62–74 (2015).
108. Iwasawa, R., Mahul-Mellier, A. L., Datler, C., 132. Mattiazzi Ušaj, M. et al. Genome-wide localization 155. Sharp, W. W. et al. Dynamin-related protein 1
Pazarentzos, E. & Grimm, S. Fis1 and Bap31 bridge study of yeast Pex11 identifies peroxisome– (Drp1)-mediated diastolic dysfunction in myocardial
the mitochondria-ER interface to establish a platform mitochondria interactions through the ERMES ischemia-reperfusion injury: therapeutic benefits
for apoptosis induction. EMBO J. 30, 556–568 complex. J. Mol. Biol. 427, 2072–2087 (2015). of Drp1 inhibition to reduce mitochondrial fission.
(2011). 133. McGuinness, M. C. et al. Role of ALDP (ABCD1) FASEB J. 28, 316–326 (2014).
109. Rusinol, A. E., Cui, Z., Chen, M. H. & Vance, J. E. and mitochondria in X-linked adrenoleukodystrophy. 156. Kim, B. et al. Inhibition of Drp1-dependent mito­
A unique mitochondria-associated membrane fraction Mol. Cell. Biol. 23, 744–753 (2003). chondrial division impairs myogenic differentiation.
from rat liver has a high capacity for lipid synthesis 134. Fan, J., Li, X., Issop, L., Culty, M. & Papadopoulos, V. Am. J. Physiol. Regul. Integr. Comp. Physiol. 305,
and contains pre-Golgi secretory proteins including ACBD2/ECI2-mediated peroxisome-mitochondria R927–R938 (2013).
nascent lipoproteins. J. Biol. Chem. 269, interactions in Leydig cell steroid biosynthesis. 157. Cereghetti, G. M., Costa, V. & Scorrano, L. Inhibition
27494–27502 (1994). Mol. Endocrinol. 30, 763–782 (2016). of Drp1-dependent mitochondrial fragmentation and
Discovery of mitochondria-associated ER membranes. 135. Kerr, J. F., Wyllie, A. H. & Currie, A. R. Apoptosis: apoptosis by a polypeptide antagonist of calcineurin.
110. Giacomello, M. & Pellegrini, L. The coming of age of a basic biological phenomenon with wide-ranging Cell Death Differ. 17, 1785–1794 (2010).
the mitochondria-ER contact: a matter of thickness. implications in tissue kinetics. Br. J. Cancer. 26, 158. Ferreira, J. C. B. et al. A selective inhibitor of mitofusin
Cell Death Differ. 23, 1417–1427 (2016). 239–257 (1972). 1-βIIPKC association improves heart failure outcome in
111. De Mario, A., Quintana-Cabrera, R., Martinvalet, D. & 136. Karbowski, M. et al. Quantitation of mitochondrial rats. Nat. Commun. 10, 329 (2019).
Giacomello, M. (Neuro)degenerated mitochondria-ER dynamics by photolabeling of individual organelles 159. Leboucher, Guillaume P. et al. Stress-induced
contacts. Biochem. Biophys. Res. Commun. 483, shows that mitochondrial fusion is blocked during the phosphorylation and proteasomal degradation of
1096–1109 (2017). Bax activation phase of apoptosis. J. Cell Biol. 164, mitofusin 2 facilitates mitochondrial fragmentation
112. Friedman, J. R., Webster, B. M., Mastronarde, D. N., 493–499 (2004). and apoptosis. Mol. Cell 47, 547–557 (2012).
Verhey, K. J. & Voeltz, G. K. ER sliding dynamics and 137. Wasiak, S., Zunino, R. & McBride, H. M. Bax/Bak 160. Shutt, T., Geoffrion, M., Milne, R. & McBride, H. M.
ER-mitochondrial contacts occur on acetylated promote sumoylation of DRP1 and its stable The intracellular redox state is a core determinant
microtubules. J. Cell Biol. 190, 363–375 (2010). association with mitochondria during apoptotic of mitochondrial fusion. EMBO Rep. 13, 909–915
113. Korobova, F., Ramabhadran, V. & Higgs, H. N. An cell death. J. Cell Biol. 177, 439–450 (2007). (2012).
actin-dependent step in mitochondrial fission 138. Braschi, E., Zunino, R. & McBride, H. M. MAPL is a 161. Mattie, S., Riemer, J., Wideman, J. G. & McBride, H. M.
mediated by the ER-associated formin INF2. Science new mitochondrial SUMO E3 ligase that regulates A new mitofusin topology places the redox-regulated
339, 464–467 (2013). mitochondrial fission. EMBO Rep. 10, 748–754 C terminus in the mitochondrial intermembrane space.
114. Manor, U. et al. A mitochondria-anchored isoform (2009). J. Cell Biol. 217, 507–515 (2018).
of the actin-nucleating spire protein regulates 139. Figueroa-Romero, C. et al. SUMOylation of the This identification of a revised topology for
mitochondrial division. eLife 4, e08828 (2015). mitochondrial fission protein Drp1 occurs at multiple mitofusin with the coiled coil 2 region exposed
115. Chakrabarti, R. et al. INF2-mediated actin nonconsensus sites within the B domain and is linked to the intermembrane space has had profound
polymerization at the ER stimulates mitochondrial to its activity cycle. FASEB J. 23, 3917–3927 (2009). implications for the mechanism of mitochondrial
calcium uptake, inner membrane constriction, and 140. Prudent, J. et al. MAPL SUMOylation of Drp1 fusion and the development of mitofusin-targeting
division. J. Cell Biol. 217, 251–268 (2017). stabilizes an ER/mitochondrial platform required for drugs.
116. Wong, L. H., Gatta, A. T. & Levine, T. P. Lipid transfer cell death. Mol. Cell 59, 941–955 (2015). 162. Cogliati, S., Enriquez, J. A. & Scorrano, L. Mitochondrial
proteins: the lipid commute via shuttles, bridges 141. Guo, X. et al. Inhibition of mitochondrial fragmentation cristae: where beauty meets functionality. Trends
and tubes. Nat. Rev. Mol. Cell Biol. 20, 85–101 diminishes Huntington’s disease-associated Biochem. Sci. 41, 261–273 (2016).
(2019). neurodegeneration. J. Clin. Invest. 123, 5371–5388 163. Yamaguchi, R. et al. Opa1-mediated cristae opening is
117. Janer, A. et al. SLC25A46 is required for (2013). Bax/Bak and BH3 dependent, required for apoptosis,
mitochondrial lipid homeostasis and cristae 142. Qi, X., Qvit, N., Su, Y. C. & Mochly-Rosen, D. A novel and independent of Bak oligomerization. Mol. Cell 31,
maintenance and is responsible for Leigh syndrome. Drp1 inhibitor diminishes aberrant mitochondrial 557–569 (2008).
EMBO Mol. Med. 8, 1019–1038 (2016). fission and neurotoxicity. J. Cell Sci. 126, 789–802 164. Chen, X. et al. Targeting mitochondrial structure
118. Wu, M. J. et al. Epithelial-mesenchymal transition (2013). sensitizes acute myeloid leukemia to venetoclax
directs stem cell polarity via regulation of mitofusin. 143. Merrill, R. A., Slupe, A. M. & Strack, S. N-terminal treatment. Cancer Discov. 9, 890–909 (2019).
Cell Metab. 29, 993–1002.e6 (2019). phosphorylation of protein phosphatase-2A/Bβ2 165. Wu, W. et al. OPA1 overexpression ameliorates
119. Singaravelu, K. et al. Mitofusin 2 Regulates STIM1 regulates translocation to mitochondria, mitochondrial cristae remodeling, mitochondrial
migration from the Ca2+ store to the plasma dynamin-related protein 1 dephosphorylation, and dysfunction, and neuronal apoptosis in prion
membrane in cells with depolarized mitochondria. neuronal survival. FEBS J. 280, 662–673 (2013). diseases. Cell Death Dis. 10, 710–710 (2019).
J. Biol. Chem. 286, 12189–12201 (2011). 144. Costa, V. et al. Mitochondrial fission and cristae 166. Varanita, T. et al. The OPA1-dependent mitochondrial
120. Ping, H. A., Kraft, L. M., Chen, W., Nilles, A. E. & disruption increase the response of cell models of cristae remodeling pathway controls atrophic,
Lackner, L. L. Num1 anchors mitochondria to the Huntington’s disease to apoptotic stimuli. EMBO Mol. apoptotic, and ischemic tissue damage. Cell Metab.
plasma membrane via two domains with different lipid Med. 2, 490–503 (2010). 21, 834–844 (2015).

Nature Reviews | Molecular Cell Biology


Reviews

167. Ramonet, D. et al. Optic atrophy 1 mediates mitochondrial network: new implications for 212. Szabadkai, G. et al. Drp-1-dependent division of the
mitochondria remodeling and dopaminergic Charcot-Marie-Tooth disease. J. Cell Biol. 170, mitochondrial network blocks intraorganellar Ca2+
neurodegeneration linked to complex I deficiency. 1067–1078 (2005). waves and protects against Ca2+-mediated apoptosis.
Cell Death Differ. 20, 77–85 (2012). 190. Neuspiel, M. et al. Cargo-selected transport from the Mol. Cell 16, 59–68 (2004).
168. Faccenda, D. et al. Control of mitochondrial mitochondria to peroxisomes is mediated by vesicular 213. Herranz, N. & Gil, J. Mitochondria and senescence:
remodeling by the ATPase inhibitory factor 1 carriers. Curr. Biol. 18, 102–108 (2008). new actors for an old play. EMBO J. 35, 701–702
unveils a pro-survival relay via OPA1. Cell Rep. 191. Hughes, A. L., Hughes, C. E., Henderson, K. A., (2016).
18, 1869–1883 (2017). Yazvenko, N. & Gottschling, D. E. Selective sorting and 214. Chapman, J., Fielder, E. & Passos, J. F. Mitochondrial
169. Sun, M. G. et al. Correlated three-dimensional light destruction of mitochondrial membrane proteins in dysfunction and cell senescence: deciphering a
and electron microscopy reveals transformation of aged yeast. eLife 5, e13943 (2016). complex relationship. FEBS Lett. 593, 1566–1579
mitochondria during apoptosis. Nat. Cell Biol. 9, 192. Mitchell, P. & Moyle, J. Chemiosmotic hypothesis of (2019).
1057–1065 (2007). oxidative phosphorylation. Nature 213, 137–139 215. Vasileiou, P. V. S. et al. Mitochondrial homeostasis
170. Otera, H., Miyata, N., Kuge, O. & Mihara, K. Drp1- (1967). and cellular senescence. Cells 8, E686 (2019).
dependent mitochondrial fission via MiD49/51 is 193. Patten, D. A. et al. OPA1-dependent cristae 216. Tezze, C. et al. Age-associated loss of OPA1 in muscle
essential for apoptotic cristae remodeling. J. Cell Biol. modulation is essential for cellular adaptation impacts muscle mass, metabolic homeostasis, systemic
212, 531–544 (2016). to metabolic demand. EMBO J. 33, 2676–2691 inflammation, and epithelial senescence. Cell Metab.
171. Germain, M., Mathai, J. P., McBride, H. M. & (2014). 25, 1374–1389 (2017).
Shore, G. C. Endoplasmic reticulum BIK initiates 194. Miyamoto, T. et al. Compartmentalized AMPK 217. Pereira, R. O. et al. OPA1 deficiency promotes
DRP1-regulated remodelling of mitochondrial cristae signaling illuminated by genetically encoded molecular secretion of FGF21 from muscle that prevents obesity
during apoptosis. EMBO J. 24, 1546–1556 (2005). sensors and actuators. Cell Rep. 11, 657–670 (2015). and insulin resistance. EMBO J. 36, 2126–2145
172. Bernardi, P., Rasola, A., Forte, M. & Lippe, G. The 195. Chouchani, E. T. et al. A unifying mechanism for (2017).
mitochondrial permeability transition pore: channel mitochondrial superoxide production during 218. Rodriguez-Nuevo, A. et al. Mitochondrial DNA
formation by F-ATP synthase, integration in signal ischemia-reperfusion injury. Cell Metab. 23, 254–263 and TLR9 drive muscle inflammation upon Opa1
transduction, and role in pathophysiology. Physiol. (2016). deficiency. EMBO J. 37, e96553 (2018).
Rev. 95, 1111–1155 (2015). 196. Zhou, H. et al. NR4A1 aggravates the cardiac 219. Restelli, L. M. et al. Neuronal mitochondrial
173. Samant, S. A. et al. SIRT3 deacetylates and activates microvascular ischemia reperfusion injury through dysfunction activates the integrated stress response
OPA1 to regulate mitochondrial dynamics during suppressing FUNDC1-mediated mitophagy and to induce fibroblast growth factor 21. Cell Rep. 24,
stress. Mol. Cell. Biol. 34, 807–819 (2014). promoting Mff-required mitochondrial fission by CK2α. 1407–1414 (2018).
174. Bossy, B. et al. S-Nitrosylation of DRP1 does not affect Basic Res. Cardiol. 113, 23 (2018). 220. Dogan, S. A. et al. Tissue-specific loss of DARS2
enzymatic activity and is not specific to Alzheimer’s 197. Mitra, K., Wunder, C., Roysam, B., Lin, G. & activates stress responses independently of
disease. J. Alzheimers. Dis. 20, S513–S526 (2010). Lippincott-Schwartz, J. A hyperfused mitochondrial respiratory chain deficiency in the heart. Cell Metab.
175. Makino, A. et al. Regulation of mitochondrial state achieved at G1-S regulates cyclin E buildup and 19, 458–469 (2014).
morphology and function by O-GlcNAcylation in entry into S phase. Proc. Natl Acad. Sci. USA 106, 221. Schneeberger, M. et al. Mitofusin 2 in POMC neurons
neonatal cardiac myocytes. Am. J. Physiol. Regul. 11960–11965 (2009). connects ER stress with leptin resistance and energy
Integr. Comp. Physiol. 300, R1296–R1302 (2011). 198. Taguchi, N., Ishihara, N., Jofuku, A., Oka, T. & imbalance. Cell 155, 172–187 (2013).
176. Pickles, S., Vigié, P. & Youle, R. J. Mitophagy and Mihara, K. Mitotic phosphorylation of dynamin-related 222. Mancini, G. et al. Mitofusin 2 in mature adipocytes
quality control mechanisms in mitochondrial GTPase Drp1 participates in mitochondrial fission. controls adiposity and body weight. Cell Rep. 26,
maintenance. Curr. Biol. 28, R170–R185 (2018). J. Biol. Chem. 282, 11521–11529 (2007). 2849–2858.e2844 (2019).
177. Mottis, A., Jovaisaite, V. & Auwerx, J. The This article identifies mitochondrial fission 223. Chung, K.-P. et al. Mitofusins regulate lipid
mitochondrial unfolded protein response in mediated by DRP1 as a crucial step in mitosis. metabolism to mediate the development of
mammalian physiology. Mamm. Genome 25, 199. Chung, J. Y., Steen, J. A. & Schwarz, T. L. lung fibrosis. Nat. Commun. 10, 3390 (2019).
424–433 (2014). Phosphorylation-induced motor shedding is required 224. Franco, A. et al. Correcting mitochondrial fusion by
178. Anand, R., Langer, T. & Baker, M. J. Proteolytic control at mitosis for proper distribution and passive manipulating mitofusin conformations. Nature 540,
of mitochondrial function and morphogenesis. inheritance of mitochondria. Cell Rep. 16, 74–79 (2016).
Biochim. Biophys. Acta 1833, 195–204 (2013). 2142–2155 (2016). Identification of the first pharmacological
179. Sugiura, A., McLelland, G. L., Fon, E. A. & 200. Kaplon, J., van Dam, L. & Peeper, D. Two-way mitochondrial fusion activators.
McBride, H. M. A new pathway for mitochondrial communication between the metabolic and cell cycle 225. Cassidy-Stone, A. et al. Chemical inhibition of the
quality control: mitochondrial-derived vesicles. machineries: the molecular basis. Cell Cycle 14, mitochondrial division dynamin reveals its role in
EMBO J. 33, 2142–2156 (2014). 2022–2032 (2015). Bax/Bak-dependent mitochondrial outer membrane
Discovery and characterization of the role of MDVs 201. Kashatus, D. F. et al. RALA and RALBP1 regulate permeabilization. Dev. Cell 14, 193–204 (2008).
in quality control of mitochondria. mitochondrial fission at mitosis. Nat. Cell Biol. 13, 226. Mallat, A. et al. Discovery and characterization of
180. Eisner, V., Picard, M. & Hajnóczky, G. Mitochondrial 1108 (2011). selective small molecule inhibitors of the mammalian
dynamics in adaptive and maladaptive cellular stress 202. Montemurro, C. et al. Cell cycle-related metabolism mitochondrial division dynamin, DRP1. Biochem.
responses. Nat. Cell Biol. 20, 755–765 (2018). and mitochondrial dynamics in a replication-competent Biophys. Res. Commun. 499, 556–562 (2018).
181. Ziviani, E., Tao, R. N. & Whitworth, A. J. Drosophila pancreatic beta-cell line. Cell Cycle 16, 2086–2099 227. Quirós, P. M., Mottis, A. & Auwerx, J. Mitonuclear
parkin requires PINK1 for mitochondrial translocation (2017). communication in homeostasis and stress. Nat. Rev.
and ubiquitinates mitofusin. Proc. Natl Acad. Sci. USA 203. Wang, H. et al. Parkin ubiquitinates Drp1 for Mol. Cell Biol. 17, 213–226 (2016).
107, 5018–5023 (2010). proteasome-dependent degradation: implication of 228. Anderson, G. R. et al. Dysregulation of mitochondrial
This article demonstrates that parkin ubiquitylates dysregulated mitochondrial dynamics in Parkinson dynamics proteins are a targetable feature of human
the mitochondrial fusion protein mitofusin, disease. J. Biol. Chem. 286, 11649–11658 (2011). tumors. Nat. Commun. 9, 1677 (2018).
providing a first mechanistic link between 204. Horn, S. R. et al. Regulation of mitochondrial 229. Antonny, B. et al. Membrane fission by dynamin:
mitophagy and mitochondrial morphology. morphology by APC/CCdh1-mediated control of Drp1 what we know and what we need to know. EMBO J.
182. Kitada, T. et al. Mutations in the parkin gene cause stability. Mol. Biol. Cell 22, 1207–1216 (2011). 35, 2270–2284 (2016).
autosomal recessive juvenile parkinsonism. Nature 205. Zunino, R., Schauss, A., Rippstein, P., 230. Chappie, J. S. et al. An intramolecular signaling
392, 605–608 (1998). Andrade-Navarro, M. & McBride, H. M. The SUMO element that modulates dynamin function in vitro
183. Narendra, D., Tanaka, A., Suen, D. F. & Youle, R. J. protease SENP5 is required to maintain mitochondrial and in vivo. Mol. Biol. Cell 20, 3561–3571 (2009).
Parkin is recruited selectively to impaired morphology and function. J. Cell Sci. 120, 231. Daumke, O., Roux, A. & Haucke, V. BAR domain
mitochondria and promotes their autophagy. 1178–1188 (2007). scaffolds in dynamin-mediated membrane fission.
J. Cell Biol. 183, 795–803 (2008). 206. Zunino, R., Braschi, E., Xu, L. & McBride, H. M. Cell 156, 882–892 (2014).
This article demonstrates that parkin translocates Translocation of SenP5 from the nucleoli to the 232. Sochacki, K. A. & Taraska, J. W. From flat to curved
to dysfunctional mitochondria, providing the first mitochondria modulates DRP1-dependent fission clathrin: controlling a plastic ratchet. Trends Cell Biol.
mechanistic explanation for selective autophagy. during mitosis. J. Biol. Chem. 284, 17783–17795 29, 241–256 (2019).
184. Park, J., Lee, G. & Chung, J. The PINK1-parkin (2009). 233. Ford, M. G., Jenni, S. & Nunnari, J. The crystal
pathway is involved in the regulation of mitochondrial 207. Zhong, X. et al. Mitochondrial dynamics is critical for structure of dynamin. Nature 477, 561–566
remodeling process. Biochem. Biophys. Res. Commun. the full pluripotency and embryonic developmental (2011).
378, 518–523 (2009). potential of pluripotent stem cells. Cell Metab. 29, 234. Roux, A., Uyhazi, K., Frost, A. & De Camilli, P.
185. Sarraf, S. A. et al. Landscape of the 979–992.e4 (2019). GTP-dependent twisting of dynamin implicates
PARKIN-dependent ubiquitylome in response to 208. Luchsinger, L. L., de Almeida, M. J., Corrigan, D. J., constriction and tension in membrane fission. Nature
mitochondrial depolarization. Nature 496, 372 Mumau, M. & Snoeck, H. W. Mitofusin 2 maintains 441, 528–531 (2006).
(2013). haematopoietic stem cells with extensive lymphoid 235. Rismanchi, N., Soderblom, C., Stadler, J., Zhu, P. P. &
186. Sugiura, A. et al. MITOL regulates endoplasmic potential. Nature 529, 528–531 (2016). Blackstone, C. Atlastin GTPases are required for Golgi
reticulum-mitochondria contacts via mitofusin2. 209. Jung, S. et al. Mitofusin 2, a mitochondria-ER apparatus and ER morphogenesis. Hum. Mol. Genet.
Mol. Cell 51, 20–34 (2013). tethering protein, facilitates osteoclastogenesis 17, 1591–1604 (2008).
187. Chen, Y. & Dorn, G. W. II PINK1-phosphorylated by regulating the calcium-calcineurin-NFATc1 axis. 236. Smith, J. J. & Aitchison, J. D. Peroxisomes take shape.
mitofusin 2 is a parkin receptor for culling damaged Biochem. Biophys. Res. Commun. 516, 202–208 Nat. Rev. Mol. Cell Biol. 14, 803–817 (2013).
mitochondria. Science 340, 471–475 (2013). (2019). 237. McNew, J. A., Sondermann, H., Lee, T., Stern, M.
188. Toyama, E. Q. et al. AMP-activated protein kinase 210. Bahat, A. et al. MTCH2-mediated mitochondrial fusion & Brandizzi, F. GTP-dependent membrane fusion.
mediates mitochondrial fission in response to energy drives exit from naïve pluripotency in embryonic stem Annu. Rev. Cell Dev. Biol. 29, 529–550 (2013).
stress. Science 351, 275–281 (2016). cells. Nat. Commun. 9, 5132 (2018). 238. Zuchner, S. et al. Mutations in the pleckstrin homology
189. Niemann, A., Ruegg, M., La Padula, V., Schenone, A. 211. Favaro, G. et al. DRP1-mediated mitochondrial domain of dynamin 2 cause dominant intermediate
& Suter, U. Ganglioside-induced differentiation shape controls calcium homeostasis and muscle mass. Charcot-Marie-Tooth disease. Nat. Genet. 37,
associated protein 1 is a regulator of the Nat. Commun. 10, 2576 (2019). 289–294 (2005).

www.nature.com/nrm
Reviews

239. Rizzuto, R. et al. Close contacts with the endoplasmic 255. Gomez-Suaga, P. et al. The ER-mitochondria tethering for OPA1 mutations. Mol. Genet. Metab. 103,
reticulum as determinants of mitochondrial Ca2+ complex VAPB-PTPIP51 regulates autophagy. Curr. 383–387 (2011).
responses. Science 280, 1763–1766 (1998). Biol. 27, 371–385 (2017). 273. Spiegel, R. et al. Fatal infantile mitochondrial
240. Giacomello, M. et al. Ca2+ hot spots on the 256. Kirmiz, M., Vierra, N. C., Palacio, S. & Trimmer, J. S. encephalomyopathy, hypertrophic cardiomyopathy
mitochondrial surface are generated by Ca2+ Identification of VAPA and VAPB as Kv2 and optic atrophy associated with a homozygous
mobilization from stores, but not by activation of channel-interacting proteins defining endoplasmic OPA1 mutation. J. Med. Genet. 53, 127–131 (2016).
store-operated Ca2+ channels. Mol. Cell 38, 280–290 reticulum-plasma membrane junctions in mammalian 274. Carelli, V. et al. Syndromic parkinsonism and dementia
(2010). brain neurons. J. Neurosci. 38, 7562–7584 (2018). associated with OPA1 missense mutations. Ann. Neurol.
241. Csordas, G. et al. Imaging interorganelle contacts 257. Hamasaki, M. et al. Autophagosomes form at 78, 21–38 (2015).
and local calcium dynamics at the ER-mitochondrial ER-mitochondria contact sites. Nature 495, 389–393 275. Harder, Z., Zunino, R. & McBride, H. Sumo1 conjugates
interface. Mol. Cell 39, 121–132 (2010). (2013). mitochondrial substrates and participates in
242. Rizzuto, R., De, S. D., Raffaello, A. & Mammucari, C. 258. Cerqua, C. et al. Trichoplein/mitostatin regulates mitochondrial fission. Curr. Biol. 14, 340–345 (2004).
Mitochondria as sensors and regulators of calcium endoplasmic reticulum-mitochondria juxtaposition. 276. Yonashiro, R. et al. A novel mitochondrial ubiquitin
signalling. Nat. Rev. Mol. Cell Biol. 13, 566–578 EMBO Rep. 11, 854–860 (2010). ligase plays a critical role in mitochondrial dynamics.
(2012). 259. Dadsena, S. et al. Ceramides bind VDAC2 to trigger EMBO J. 25, 3618–3626 (2006).
243. Wu, S. et al. Binding of FUN14 domain containing 1 mitochondrial apoptosis. Nat. Commun. 10, 1832 277. Burchell, V. S. et al. The Parkinson’s disease–linked
with inositol 1,4,5-trisphosphate receptor in (2019). proteins Fbxo7 and parkin interact to mediate
mitochondria-associated endoplasmic reticulum 260. Soubannier, V. et al. A vesicular transport pathway mitophagy. Nat. Neurosci. 16, 1257 (2013).
membranes maintains mitochondrial dynamics and shuttles cargo from mitochondria to lysosomes. 278. Park, Y.-Y. et al. Loss of MARCH5 mitochondrial E3
function in hearts in vivo. Circulation 136, Curr. Biol. 22, 135–141 (2012). ubiquitin ligase induces cellular senescence through
2248–2266 (2017). 261. Magni, G., Emanuelli, M., Amici, A., Raffaelli, N. & dynamin-related protein 1 and mitofusin 1. J. Cell Sci.
244. Park, S. J. et al. DISC1 modulates neuronal stress Ruggieri, S. Purification of human nicotinamide- 123, 619–626 (2010).
responses by gate-keeping er-mitochondria Ca2+ mononucleotide adenylyltransferase. Methods 279. Pyakurel, A., Savoia, C., Hess, D. & Scorrano, L.
transfer through the MAM. Cell Rep. 21, 2748–2759 Enzymol. 280, 241–247 (1997). Extracellular regulated kinase phosphorylates
(2017). 262. Soubannier, V., Rippstein, P., Kaufman, B. A., mitofusin 1 to control mitochondrial morphology
245. Simmen, T. et al. PACS-2 controls endoplasmic Shoubridge, E. A. & McBride, H. M. Reconstitution of and apoptosis. Mol. Cell 58, 244–254 (2015).
reticulum–mitochondria communication and mitochondria derived vesicle formation demonstrates 280. Zhou, W. et al. Mutation of the protein kinase a
Bid-mediated apoptosis. EMBO J. 24, 717–729 selective enrichment of oxidized cargo. PLoS One 7, phosphorylation site influences the anti-proliferative
(2005). e52830 (2012). activity of mitofusin 2. Atherosclerosis 211, 216–223
246. Vance, J. E. Newly made phosphatidylserine and 263. McLelland, G. L., Soubannier, V., Chen, C. X., (2010).
phosphatidylethanolamine are preferentially McBride, H. M. & Fon, E. A. Parkin and PINK1 281. Nakamura, N., Kimura, Y., Tokuda, M., Honda, S.
translocated between rat liver mitochondria and function in a vesicular trafficking pathway regulating & Hirose, S. MARCH-V is a novel mitofusin 2- and
endoplasmic reticulum. J. Biol. Chem. 266, 89–97 mitochondrial quality control. EMBO J. 33, 282–295 Drp1-binding protein able to change mitochondrial
(1991). (2014). morphology. EMBO Rep. 7, 1019–1022 (2006).
247. Naon, D. et al. Critical reappraisal confirms that 264. Waterham, H. R. et al. A lethal defect of mitochondrial 282. Song, Z., Chen, H., Fiket, M., Alexander, C. & Chan, D.
mitofusin 2 is an endoplasmic reticulum-mitochondria and peroxisomal fission. N. Engl. J. Med. 356, C. OPA1 processing controls mitochondrial fusion and
tether. Proc. Natl Acad. Sci. USA 113, 11249–11254 1736–1741 (2007). is regulated by mRNA splicing, membrane potential,
(2016). 265. Gerber, S. et al. Mutations in DNM1L, as in OPA1, and Yme1L. J. Cell Biol. 178, 749–755 (2007).
248. Chen, Y. et al. Mitofusin 2-containing mitochondrial- result in dominant optic atrophy despite opposite
reticular microdomains direct rapid cardiomyocyte effects on mitochondrial fusion and fission. Brain 140, Acknowledgements
bioenergetic responses via interorganelle Ca2+ 2586–2596 (2017). Research in the L.S. laboratory is supported by AIRC
crosstalk. Circ. Res. 111, 863–875 (2012). 266. Sheffer, R. et al. Postnatal microcephaly and pain IG19991, Italian Ministry of Education, University and
249. Bassoy, E. Y. et al. ER-mitochondria contacts control insensitivity due to a de novo heterozygous DNM1L Research PRIN 2017BF3PXZ, Fondation Leducq TNE15004,
surface glycan expression and sensitivity to killer mutation causing impaired mitochondrial fission and Muscular Dystrophy Association RG 603731 and Cariparo
lymphocytes in glioma stem-like cells. EMBO J. 36, function. Am. J. Med. Genet. A 170, 1603–1607 Foundation SIGMI. Research in the M.G. laboratory is sup-
1493–1512 (2017). (2016). ported by CARIPARO Starting Grant 2016 AIFbiol and Unipd
250. Naon, D. et al. Does mitofusin 2 tether or separate 267. Baxter, R. V. et al. Ganglioside-induced differentiation- STARS Consolidator FIRMESs.
endoplasmic reticulum and mitochondria? reply. associated protein-1 is mutant in Charcot-Marie-Tooth
Proc. Natl Acad. Sci. USA 114, E2268–E2269 disease type 4A/8q21. Nat. Genet. 30, 21–22 (2002). Author contributions
(2017). 268. Boyer, O. et al. INF2 mutations in Charcot–Marie– M.G. and L.S. conceptualized, wrote most of and edited the
251. Cosson, P., Marchetti, A., Ravazzola, M. & Orci, L. Tooth disease with glomerulopathy. N. Engl. J. Med. article. A.P. and C.G. wrote subsections. All authors approved
Mitofusin-2 independent juxtaposition of endoplasmic 365, 2377–2388 (2011). the final content.
reticulum and mitochondria: an ultrastructural study. 269. Brown, E. J. et al. Mutations in the formin gene INF2
PLoS One 7, e46293 (2012). cause focal segmental glomerulosclerosis. Nat. Genet. Competing interests
252. Filadi, R. et al. Mitofusin 2 ablation increases 42, 72 (2009). The authors declare no competing interests.
endoplasmic reticulum-mitochondria coupling. 270. Koch, J. et al. Disturbed mitochondrial and peroxisomal
Proc. Natl Acad. Sci. USA 112, E2174–E2181 (2015). dynamics due to loss of MFF causes Leigh-like Publisher’s note
253. Bravo, R. et al. Increased ER-mitochondrial coupling encephalopathy, optic atrophy and peripheral Springer Nature remains neutral with regard to jurisdictional
promotes mitochondrial respiration and bioenergetics neuropathy. J. Med. Genet. 53, 270–278 (2016). claims in published maps and institutional affiliations.
during early phases of ER stress. J. Cell Sci. 124, 271. Shamseldin, H. E. et al. Genomic analysis of
2143–2121s2152 (2011). mitochondrial diseases in a consanguineous Supplementary information
254. De Vos, K. J. et al. VAPB interacts with the population reveals novel candidate disease genes. Supplementary information is available for this paper at
mitochondrial protein PTPIP51 to regulate calcium J. Med. Genet. 49, 234–241 (2012). https://doi.org/10.1038/s41580-020-0210-7.
homeostasis. Hum. Mol. Genet. 21, 1299–1311 272. Schaaf, C. P. et al. Early-onset severe neuromuscular
(2012). phenotype associated with compound heterozygosity © Springer Nature Limited 2020

Nature Reviews | Molecular Cell Biology

You might also like