Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Biophysical Reviews

https://doi.org/10.1007/s12551-021-00891-w

REVIEW

Uncovering the important role of mitochondrial dynamics


in oogenesis: impact on fertility and metabolic disorder transmission
Marcos Roberto Chiaratti1

Received: 5 September 2021 / Accepted: 1 November 2021


© International Union for Pure and Applied Biophysics (IUPAB) and Springer-Verlag GmbH Germany, part of Springer Nature 2021

Abstract
Oocyte health is tightly tied to mitochondria given their role in energy production, metabolite supply, calcium (­ Ca2+) buffer-
ing, and cell death regulation, among others. In turn, mitochondrial function strongly relies on these organelle dynamics once
cyclic events of fusion and fission (division) are required for mitochondrial turnover, positioning, content homogenization,
metabolic flexibility, interaction with subcellular compartments, etc. Importantly, during oogenesis, mitochondria change
their architecture from an “orthodox” elongated shape characterized by the presence of numerous transversely oriented
cristae to a round-to-oval morphology containing arched and concentrically arranged cristae. This, along with evidence
showing that mitochondrial function is kept quiescent during most part of oocyte development, suggests an important role
of mitochondrial dynamics in oogenesis. To investigate this, recent works have downregulated/upregulated in oocytes the
expression of key effectors of mitochondrial dynamics, including mitofusins 1 (MFN1) and 2 (MFN2) and the dynamin-
related protein 1 (DRP1). As a result, both MFN1 and DRP1 were found to be essential to oogenesis and fertility, while MFN2
deletion led to offspring with increased weight gain and glucose intolerance. Curiously, neither MFN1/MFN2 deficiency
nor DRP1 overexpression enhanced mitochondrial fragmentation, indicating that mitochondrial size is strictly regulated
in oocytes. Therefore, the present work seeks to discuss the role of mitochondria in supporting oogenesis as well as recent
findings connecting defective mitochondrial dynamics in oocytes with infertility and transmission of metabolic disorders.

Keywords Oocyte · Mitochondria · Endoplasmic reticulum · MFN1 · MFN2 · DRP1

Introduction et al. 2008). The mitochondrion presents in this context as a


pivotal organelle due to its key role in apoptosis and energy
In humans, the ovary is one of the first organs to fail with production for normal spindle assembly and chromosomal
aging due to a progressive reduction of both follicle (the segregation (Al-Zubaidi et al. 2021; Babayev et al. 2016;
functional unit of the ovary) number and egg quality. Con- Bentov et al. 2011; Johnson et al. 2007; May-Panloup et al.
sequently, live birth rate declines from 43%, among women 2016; Pasquariello et al. 2019; Tilly and Sinclair, 2013;
younger than 35 years, to 6%, among women aged 42 years Van Blerkom et al. 1995, 2000; Yu et al. 2010). In addition,
or more (Baird et al. 2005; Schwartz and Mayaux, 1982; numerous metabolic processes regulated by mitochondria
Van Noord-Zaadstra et al. 1991; Wright et al. 2008). While such as calcium (­ Ca2+) buffering, phospholipid biosynthe-
the decline in follicle number is thought to be explained sis, and reduction–oxidation state can affect egg formation
by increased follicle atresia as a result of programed cell (Al-Zubaidi et al. 2021; Carroll et al. 1994; May-Panloup
death (apoptosis), meiotic errors resulting in aneuploidy et al. 2016; Spinelli and Haigis, 2018). In face of these find-
are the leading cause of poor egg quality (Schwartz and ings, mitochondrial quality and number have been proposed
Mayaux, 1982; Van Noord-Zaadstra et al. 1991; Wright as markers of egg quality. Indeed, there are several reports
showing that mitochondrial DNA (mtDNA) copy number
can be highly variable among oocytes (and, consequently,
* Marcos Roberto Chiaratti
marcos.chiaratti@ufscar.br embryos) and associate with developmental potential,
although further studies are needed to verify the extension
1
Departamento de Genética e Evolução, Centro de Ciências and reliability of this association (Diez-Juan et al. 2015; Fra-
Biológicas e da Saúde, Universidade Federal de São Carlos, gouli et al. 2015; Pasquariello et al. 2019; Santos et al. 2006;
São Carlos 13565‑905, Brazil

13
Vol.:(0123456789)
Biophysical Reviews

Seli 2016; Wai et al. 2010). Moreover, several attempts have form a nucleoid, a likely platform for replication, transcrip-
been made to rescue poor quality eggs through supplemen- tion, and translation of mtDNA (reviewed by (Gustafsson
tation with exogenous mitochondria, being their results a et al. 2016)). Due to its relative short half-life, mtDNA is
matter of constant debates (Cohen et al. 1997; Kristensen replicated at a correspondingly high rate, making it prone
et al. 2017; Labarta et al. 2019a, b; Woods and Tilly, 2015). to replicative errors (Krasich and Copeland, 2017). As such,
The mitochondrial network can vary from hundreds to mutated mtDNA can coexist in a cell with wild-type mol-
millions of mitochondria depending on the cell type and ecules (referred to as heteroplasmy), potentially leading to
on the balance between mitochondrial fusion and fission devastating diseases (reviewed by (Craven et al. 2017)).
(division). In turn, each mitochondrion is composed of two The present work aims to review the current knowledge
membranes, the inner and the outer, which delimitate two regarding the role of mitochondria in supporting oocyte
compartments, the mitochondrial lumen (matrix) and the development. Several works encompassing morphofunc-
intermembrane space; invagination of the inner membrane tional characteristics of oocyte mitochondria are discussed,
towards the matrix further determines the mitochondrial with an emphasis on recent findings connecting defective
cristae (reviewed by (Pernas and Scorrano, 2016)). Within mitochondrial dynamics in oocytes with infertility and inter-
the mitochondrial matrix resides the enzymes taking part generational transmission of metabolic disorders.
in the tricarboxylic acid cycle and β-oxidation, biochemi-
cal processes responsible for the oxidation of energetic sub-
strates. Nicotinamide adenine dinucleotide (NAD)H and The unique characteristics of mitochondria
flavin adenine dinucleotide (FAD)H2, generated from these in oocytes
oxidative processes, sustain the electron transport chain
(ETC) through four enzymatic complexes in the cristae Oogenesis commences with specification of primordial germ
membrane, enabling reduction of oxygen ­(O2) into water cells (PGCs) in the posterior epiblast base in pre-gastrula-
as well as the formation of a proton ­(H+) gradient across tion post-implantation embryos. Thereafter, PGCs migrate
the inner mitochondrial membrane. Eventually, the ­H+ gra- to the gonadal ridges, differentiate into oogonia, and pro-
dient is used by a fifth enzymatic complex (the F ­ 1F0-ATP liferate by mitosis prior to undergoing meiosis and giving
synthase) in the cristae membrane to phosphorylate ADP rise to oocytes (reviewed by (Kobayashi and Surani, 2018)).
into ATP, this latter representing the most abundant energy Oocytes progress through the first stages of meiosis until
source for biological processes in the cell. In addition to reaching the late-diplotene resting stage referred to as dicty-
ATP, reactive oxygen species (ROS) such as hydrogen ate; meiosis is resumed only after puberty, which in humans
peroxide, superoxide, and hydroxyl radicals can be gener- can take 40 years or more. At birth, the ovary is therefore
ated from the oxidative phosphorylation (OXPHOS) due populated with up to hundreds of thousands of oocytes sur-
to incomplete ­O2 reduction. ROS can damage membranes, rounded by a single layer of squamous somatic granulosa
proteins, and nucleic acids, but the mitochondrion owns a cells (Fig. 1). These germ-soma structures, called primordial
highly effective mechanism for dealing with its production. follicles, remain quiescent until they are recruited for devel-
Under certain circumstances, however, ROS production sur- opment, which encompasses proliferation and differentiation
passes the mitochondrial oxidative defenses, with potential of granulosa cells along with a ~ 100-fold increase in oocyte
consequences to mitochondrial and cellular health (reviewed volume (Liu et al. 2014; Pedersen and Peters, 1968; Reddy
by (May-Panloup et al. 2016)). et al. 2008; Rimon-Dahari et al. 2016; Zhang and Liu, 2015).
The majority of the mitochondrial proteome (~ 1100 pro- During this period, known as folliculogenesis, the oocyte
teins) is encoded in the nucleus, translated in the cytoplasm, progresses through a series of changes and stockpiles sev-
and imported into mitochondria (Rath et al. 2020). None- eral molecules and organelles, events that rely on the close
theless, mitochondria have their own genome (mtDNA), interaction of the gamete with granulosa cells (Fig. 1). Even-
which in mammals is exclusively inherited from the mother, tually, the oocyte resumes meiosis and is ovulated at the
has ~ 16.5 kb, and encompasses 37 genes encoding for 13 metaphase II stage (Fig. 1), even though many oocytes can
polypeptides of the OXPHOS complexes (reviewed by be eliminated through follicle atresia (reviewed by (Binelli
(Stewart and Chinnery, 2020)). Additionally, recent works and Murphy, 2010; Clarke, 2017)).
suggest a role for mtDNA in encoding mitochondrial derived While mitochondria in PGCs have a rounded shape
peptides (MDPs) and small mitochondrial RNAs (Miller (0.4–0.6 μm in diameter) and tubulo-vesicular cristae, these
et al. 2020; Pozzi et al. 2019). Multiple mtDNA copies are organelles get enlarged (0.8–1.0 μm in diameter) after differ-
present in each cell, being mtDNA copy number closely entiating into oogonia, besides owing sparse, lamellar cristae
associated with the cell’s energy demand. One or more cop- (Motta et al. 2000). In turn, during oocyte differentiation,
ies of mtDNA are packaged together along with proteins mitochondria assume an “orthodox” (Fig. 1) configuration
such as the mitochondrial transcription factor A (TFAM) to characterized by an elongated shape (1.2–1.9 μm in length)

13
Biophysical Reviews

Primordial Primary Tertiary


follicle follicle follicle Ovulation

granulosa
cells

oocyte ZP

granulosa cell
granulosa cell
(squamous)
(cuboid)
vacuole oocyte
dumbbell-shaped
mitochondrion ER
granulosa cell-
oocyte coupling

“orthodox” rounded lipid


mitochondrion mitochondrion droplets

Fig. 1  Unique characteristics of mitochondria in oocytes. Starting cristae. Moreover, as oocytes develop, mitochondria, the smooth
with puberty, a group of primordial follicles initiates development endoplasmic reticulum (ER), and lipid droplets become more fre-
as represented by proliferation and differentiation of somatic granu- quently associated, pointing towards an important role of such orga-
losa cells as well as oocyte growth. This period, known as follicu- nelle cluster (referred to as metabolic units) in late oogenesis. These
logenesis, encompasses several stages (i.e., primary, secondary, and changes in mitochondrial morphology, along with functional data,
tertiary follicles) and eventually should result in the ovulation of a suggest that mitochondrial activity is downregulated through oogen-
gamete competent for fertilization and embryogenesis. In turn, mito- esis. In agreement with this, granulosa cells generate multiple filopo-
chondrial architecture changes dramatically during oocyte develop- dia (arrowheads in the inset) that penetrate the zona pellucida (ZP)
ment. Initially, mitochondria display an “orthodox” configuration to reach the oocyte membrane and establish gap/adherens junctions.
characterized by an elongated shape, (1.2–1.9 μm in length), numer- Apart from acting as a signaling hub, these cell junctions are used
ous transversely oriented cristae, and a single large vacuole. However, by granulosa cells to provide the gamete with energetic molecules
the increased abundance of dumbbell-shaped mitochondria in early- such as ATP, pyruvate, nicotinamide adenine dinucleotide phosphate
developing oocytes indicates a change in mitochondrial dynamics that (NADP)H, cholesterol, and certain amino acids. The inset in “Ovula-
results, near ovulation, in mitochondria that are round (0.4–0.6 μm in tion” depicts an ovulated oocyte with mitochondria stained in green
diameter), vacuolated, and have arched and concentrically arranged and visualized through confocal microscopy (scale bar = 25 μm)

and numerous transversely oriented cristae, apart from a sin- increase compared to the number in PGCs (Cao et al. 2007;
gle large vacuole (Wassarman and Josefowicz, 1978). These Jansen and De Boer, 1998; Motta et al. 2000; Wai et al.
morphological changes, which are suggestive of increased 2008; Wassarman and Josefowicz, 1978). Although this
OXPHOS capacity, are, however, transient as dumbbell- enormous population has been attributed to an essential role
shaped mitochondria become abundant in early develop- of the organelle (Harris et al. 2009), mitochondrial density
ing oocytes (Fig. 1), eventually giving rise to round/oval decreases with folliculogenesis progression (Babayev et al.
vacuolated organelles displaying columnar-shaped, arched 2016). Moreover, growing oocytes have fragmented mito-
cristae (Wassarman and Josefowicz, 1978). Following, chondria and poorly developed mitochondrial cristae and are
mitochondria in fully developed oocytes remain round/oval only slightly affected by PDHA1 deficiency (Johnson et al.
(0.4–0.6 μm in diameter), contain a large vacuole, and have 2007; Kruip et al. 1983; Motta et al. 2000; Tarazona et al.
arched/concentrically-arranged cristae (Fig. 1) (Trebichalská 2006; Trebichalská et al. 2021; Wassarman and Josefowicz,
et al. 2021; Wassarman and Josefowicz, 1978). It is worth 1978), collectively suggesting that mitochondrial activity is
noting that in ruminants such as cows, mitochondria assume likely downregulated in oocytes. Indeed, there are several
a singular hooded morphology in oocytes, with unclear func- reports showing that both pyruvate consumption (normal-
tional consequences (Fair et al. 1997). Moreover, as oocytes ized by oocyte volume) and mitochondrial-coupled oxygen
develop, mitochondria, the smooth endoplasmic reticulum consumption decrease with oocyte development (Biggers
(ER), and lipid droplets become more frequently associated et al. 1967; Eppig, 1976; Harris et al. 2009; Hashimoto
(Fig. 1), pointing towards an important role of such orga- et al. 2017; Magnusson et al. 1986; Porterfield et al. 1998;
nelle cluster (referred to as metabolic units) in late oogenesis Thompson et al. 1996; Trimarchi et al. 2000). Therefore, it
(Kruip et al. 1983; Motta et al. 2000; Trebichalská et al. is believed that the gamete is kept at a relatively quiescent
2021). catabolic status due to the action of granulosa cells in pro-
About one hundred thousand mitochondria are estimated viding the oocyte with several energetic molecules, includ-
in fully-grown oocytes, which represents a ~ 1000-fold ing ATP, pyruvate, NAD phosphate (NADP)H, cholesterol,

13
Biophysical Reviews

and certain amino acids (Fig. 1) (Biggers et al. 1967; Downs,


1995; Eppig et al. 2005; Johnson et al. 2007; Richani et al.
2021; Su et al. 2007, 2009; Sugiura et al. 2007). Accord-
ingly, reprogramming of somatic cells into a pluripotent ER
state is paralleled by a metabolic switch characterized by MERCs
fragmentation of the mitochondrial network and decreased mitochondrial fusion
mitochondrial activity (Chen and Chan, 2017; Cho et al.
2006; Ma et al. 2015a; Prigione et al. 2014; Son et al. 2015).
On the other hand, mitochondrial-coupled ATP formation is fusion
essential for resumption of meiosis, spindle formation, and
chromosome segregation, likely due to end of granulosa cell-
oocyte cooperation near ovulation (Abbassi et al. 2021; Al-
Zubaidi et al. 2019; Brevini et al. 2005; Dalton et al. 2014;
Downs, 1995; Downs et al. 2002; Krisher and Bavister,
1998; Kruip et al. 1983; Pasquariello et al. 2019; Stojkovic fission
et al. 2001; Van Blerkom et al. 1995; Yu et al. 2010). elongated fragmented
mitochondria mitochondria

Main effectors of mitochondrial dynamics mitochondrial fission


MFN2
A large body of evidence has implicated mitochondria in MFN1 cristae
a range of complex biological processes encompassing OPA1
cellular signaling, differentiation, migration, stemness, DRP1
autophagy, senescence, inflammation, immune response, matrix
and cell death, apart from their long-standing role in energy
production. Additionally, mitochondria interact with each ER
other and with subcellular compartments such as the ER,
lysosomes, peroxisomes, endosomes, melanosomes, lipid Fig. 2  Main effectors of mitochondrial dynamics. Cyclic events of
droplets, the plasma membrane, and the nuclear membrane organelle fusion and fission (division) driving, respectively, elonga-
tion and fragmentation of the mitochondrial network are responsible
(reviewed by (Gordaliza‐Alaguero et al. 2019)). To effi- for determining mitochondrial dynamics. Towards that, mitofusins 1
ciently play these functions, mitochondria are constantly (MFN1) and 2 (MFN2) as well as the optic atrophy protein 1 (OPA1)
adapting their morphology and localization in the cell, which are the main effectors of fusion. MFN1 and MFN2 are both located
is collectively known as mitochondrial dynamics (Fig. 2). in the outer mitochondrial membrane, whereas OPA1 resides in the
inner mitochondrial membrane. While MFN1 and OPA1 have a cen-
As such, the mitochondrial network can range in shape from tral role in the fusion reaction, MFN2 is a key regulator of the oxi-
isolated, rounded organelles to long interconnected tubules. dative metabolism and has been implicated on mitochondria-endo-
In addition, mitochondria can distribute evenly across the plasmic reticulum (ER) contact sites (MERCs) formation. OPA1 is
cytosol or form clusters to meet local demands, events that also central to mitochondrial cristae remodeling. In turn, mitochon-
drial fission is primarily carried out by the dynamin-related protein
are cell specific and can vary throughout the cell cycle and 1 (DRP1), which locates in the cytosol and acts at sites of mitochon-
in response to metabolic and cellular clues (reviewed by dria-ER membrane juxtaposition
(Pernas and Scorrano, 2016)).
Mitochondrial dynamics is essential for maintenance of
health organelles as repeated cycles of organelle fusion and et al. 2003, 2005, 2007, 2010). MFN1 and MFN2 are both
fission are required for homogenization of the mitochondrial located in the outer mitochondrial membrane, whereas OPA1
network (Ban-Ishihara et al. 2013; Chen et al. 2003, 2005; resides in the inner mitochondrial membrane (Fig. 2). While
Ishihara et al. 2015; Twig et al. 2008). Furthermore, mito- MFN1 and OPA1 have a central role in the fusion reaction,
chondrial dynamics regulates organelle transport, inherit- the exact role of MFN2 in fusion remains unclear. OPA1
ance, and selective removal (Kleele et al. 2021; Lieber et al. is also central to mitochondrial cristae remodeling, being
2019; Misko et al. 2012; Twig et al. 2008), with defects in its function under the control of multiple enzymes such as
this machinery leading to a range of diseases in humans YME1L1, OMA1, and PARL (reviewed by (Pernas and
(Table 1). Mitofusins 1 (MFN1) and 2 (MFN2) as well as Scorrano, 2016)). More recently, mitoguardins 1 (MIGA1)
the optic atrophy protein 1 (OPA1), all belonging to the and 2 (MIGA1), which act downstream of mitofusins, have
dynamin-related family of large GTPases, are the main effec- also been shown to regulate mitochondrial fusion by inter-
tors responsible for mitochondrial fusion in mammals (Chen acting with mitochondrial phospholipase D (MitoPLD) to

13
Table 1  Main effectors of mitochondrial dynamics and their role in diseases
Gene Main role Disease/symptoms References
Biophysical Reviews

Mitofusin 1 (MFN1) Outer mitochondrial membrane protein that Unknown Chen et al. (2003); Chen et al. (2005); Chen
mediates mitochondrial fusion et al. (2007); Chen et al. (2010)
Mitofusin 2 (MFN2) Outer mitochondrial membrane protein that Charcot-Marie-Tooth disease type 2A Brockman et al. (2008); Chen et al. (2003);
mediates mitochondrion-mitochondrion (CMTD2A) Chen et al. (2007); Chen et al. (2005); Chen
interaction and juxtaposition of mitochon- Hereditary motor and sensory neuropathy et al. (2010); Fyfe et al. (2011); Rouzier
dria with the endoplasmic reticulum (HMSN) et al. (2012); Sawyer et al. (2015); Zhu et al.
Multiple symmetric lipomatosis (MSL) (2005); Züchner et al. (2004)
Canine fetal-onset neuroaxonal dystrophy
Optic atrophy 1 protein (OPA1) Inner mitochondrial membrane protein that Autosomal dominant optic atrophy (ADOA) Alexander et al. (2000); Amati-Bonneau et al.
mediates mitochondrial fusion Mitochondrial DNA depletion syndrome (2008); Carelli et al. (2015); Delettre et al.
Behr syndrome (2000); Hudson et al. (2008); Schaaf et al.
Syndromic Parkinson disease and dementia (2011); Spiegel et al. (2016)
Yeast mitochondrial escape 1-like 1 Inner mitochondrial membrane peptidase that Intellectual disability, motor development Anand et al. (2014); Hartmann et al. (2016)
(YME1L1) mediates OPA1 processing delay, expressive speech delay, optic nerve
atrophy associated with visual impairment,
and hearing impairment
OMA1 zinc metalloprotease Inner mitochondrial membrane peptidase that Unknown Anand et al. (2014)
mediates OPA1 processing
Rbd1p orthologue presenilin-associated Inner mitochondrial membrane peptidase that Unknown Saita et al. (2017)
rhomboid-like protein (PARL) mediates OPA1 processing
Dynamin-related protein 1 (DRP1) Cytoplasmic protein that mediates mitochon- Encephalopathy Fahrner et al. (2016); Gerber et al. (2017);
drial fission Autosomal dominant optic atrophy Ishihara et al., (2009); Kleele et al., (2021);
Microcephaly and pain insensitivity Losón et al. (2013); Sheffer et al. (2016);
Intractable forms of epilepsy Vanstone et al. (2016); von Spiczak et al.
Neonatal lethality (2017); Waterham et al. (2007)
Mitochondrial fission factor (MFF) Outer mitochondrial membrane receptor that Leigh-like encephalopathy, optic atrophy, and Kleele et al., (2021); Koch et al. (2016); Losón
recruits DRP1 for organelle fission peripheral neuropathy et al. (2013); Palmer et al. (2013); Shamseldin
Encephalopathy due to defective mitochon- et al. (2012)
drial and peroxisomal fission
Mitochondrial fission 1 protein (FIS1) Outer mitochondrial membrane receptor that Unknown Kleele et al., (2021); Losón et al. (2013);
recruits DRP1 for organelle fission Palmer et al. (2013); Shen et al. (2014);
Mitochondrial dynamics protein of 49 kDa Outer mitochondrial membrane receptor that Unknown Losón et al. (2013); Palmer et al. (2013)
(MID49) recruits DRP1 for organelle fission
Mitochondrial dynamics protein of 51 kDa Outer mitochondrial membrane receptor that Unknown Losón et al. (2013); Palmer et al. (2013)
(MID51) recruits DRP1 for promoting organelle fis-
sion

13
Biophysical Reviews

facilitate MitoPLD dimer formation (Zhang et al. 2016a). period (≤ 15 h) of mitofusin overexpression or restricted to
On other hand, mitochondrial fission is primarily carried out late stages of oogenesis as mitochondrion-mitochondrion
by the dynamin-related protein 1 (DRP1), which locates in contact sites were clearly increased following mitofusin
the cytosol and is attracted to the mitochondrial outer mem- overexpression (Wakai et al. 2014). In addition, mitofusin
brane by receptors such as the mitochondrial fission factor overexpression led to markedly aggregated mitochondria,
(MFF), the mitochondrial fission 1 protein (FIS1), and the particularly around the nucleus/chromosomes (Wakai et al.
mitochondrial dynamics proteins of 49 kDa (MID49) and 2014), a phenotype that relied on MFN1 GTPase activity and
51 kDa (MID51). Once bound to these receptors, DRP1 is associated with disrupted spindle/metaphase plate organiza-
capable of oligomerizing and driving mitochondrial division tion (Wakai et al. 2014).
(Fig. 2) (Giacomello et al. 2020; Ishihara et al. 2009; Kleele Targeted deletion of DRP1 during oocyte development
et al. 2021). resulted in female infertility due to impaired folliculogen-
Prior to mitochondrial fission, the smooth ER wraps esis, a result attributed to depleted ER C­ a2+ stores, multio-
around mitochondria to possibly drive, with the aid of rganelle (i.e., mitochondria, ER, peroxisomes and vesicles)
actin filaments, initial organelle constriction (Fig. 2). Actin aggregation, and defective secretory vesicle release (Fig. 3a)
polymerization further supports DRP1 oligomerization and (Udagawa et al. 2014). Curiously, neither mitochondrial
GTP hydrolysis-mediated membrane constriction (Chakra- membrane potential nor mtDNA copy number was altered
barti et al. 2018; Korobova et al. 2013; Manor et al. 2015). in DRP1-deficient oocytes, although nucleoids were highly
It is noteworthy that MFF and DRP1 often colocalizes with clustered (Udagawa et al. 2014). The apparent lack of effect
nucleoids, thus favoring maintenance of small and frag- on mitochondrial function contrasts with reports showing
mented nucleoids, cristae structure, and the pro-apoptotic that fission is critical for mitochondrial network homogeni-
status of mitochondria (Ban-Ishihara et al. 2013; Lewis zation and turnover (Ban-Ishihara et al. 2013; Ishihara et al.
et al. 2016). Mitochondria-ER contact sites (MERCs) serve, 2015; Ota et al. 2020; Twig et al. 2008), a finding that might
therefore, as fission platforms, apart from acting as impor- be secondary to the unique characteristics of mitochondria
tant hubs for lipid trafficking, ­Ca2+ signaling, ER stress, in oocytes. In turn, targeted deletion of MFN1 strongly pre-
apoptosis, and macroautophagy. Several protein complexes vented mtDNA accumulation during oocyte growth as well
such as VAPB-PTPIP51, IP3R1/2/3-GRP75-VDAC1, and as led to evident mitochondrial dysfunction characterized by
BAP31-FIS1 have been implicated in mitochondria-ER jux- organelle swelling, abnormal expression of OXPHOS subu-
taposition. The same holds true for MFN2 (Fig. 2), which is nits, decreased membrane potential, increased FAD level,
also present in the ER membrane where from it can homo- increased ROS level, and decreased ATP content (Fig. 3b).
oligomerize and hetero-oligomerize with either MFN2 or MFN1-deficient oocytes also contained highly clustered
MFN1 in mitochondria to tether both organelle membranes mitochondria with severe ultrastructure abnormalities such
(de Brito and Scorrano, 2008; Gordaliza‐Alaguero et al. as inner membrane vesicles and decreased number of cristae
2019). (Carvalho et al. 2020; Hou et al. 2019; Zhang et al. 2019a).
These findings are consistent with previous reports show-
ing that MFN1 is required for mitochondrial health, mtDNA
Regulation of mitochondrial dynamics stability, and OPA1 activity (Chen et al. 2003, 2005, 2007,
in oocytes and implications to fertility 2010; Cipolat et al. 2004), thus confirming the important
role of fusion in oocytes as evidenced for MIGA1 and
Given that oocyte mitochondria have a fragmented morphol- MIGA2 knockouts (Liu et al. 2016b). Nonetheless, neither
ogy, are vacuolated, and contain arched and concentrically MFN1 deficiency nor DRP1 overexpression enhanced mito-
arranged cristae, it is tempting to suppose that these char- chondrial fragmentation (Carvalho et al. 2020; Hou et al.
acteristics are coordinated by specific regulation of mito- 2019; Wakai et al. 2014; Zhang et al. 2019a), indicating
chondrial dynamics in oocytes. To investigate this, Udagawa that mitochondrial size is strictly regulated in oocytes. This
et al. (2014) performed oocyte-specific deletion of DRP1 in finding contrasts, however, with that of oocytes lacking
mice, finding that oocyte mitochondria are fusion compe- the mitochondrial unfolded protein response CLPP, which
tent as fission deficiency resulted in elongated organelles did display smaller mitochondria in addition to decreased
(Fig. 3a). This conclusion is further supported by the pres- expression of profusion genes Mfn1, Mfn2, and Opa1 (Wang
ence of profusion proteins MFN1, MFN2, and OPA1 in et al. 2018a).
oocytes (Liu et al. 2016a; Wakai et al. 2014). Yet, overex- Apart from an evident mitochondrial dysfunction, MFN1-
pression of either MFN1 or MFN2 did not lead to mitochon- deficient oocytes also presented increased apoptosis, disrup-
drial tubulation (Wakai et al. 2014), supporting in oocytes tion of adherens/gap junctions, and impaired communication
an imbalance of mitochondrial dynamics towards fission. with granulosa cells (Fig. 3b) (Carvalho et al. 2020; Zhang
Alternatively, this result might be attributed to the short et al. 2019a). Remarkably, females with MFN1-deficient

13
Biophysical Reviews

a b MFN1 knockout
DRP1 knockout
mitochondrial
elongation
mitochondrial vesicles
dysfunction
mtDNA

ROS

Ca2+

ER
apoptosis PI3K-Akt ATP
multiorganelle secretory vesicle signaling
aggregation release

granulosa cell- granulosa cell-


oocyte communication oocyte communication

folliculogenesis folliculogenesis
fertility fertility

c MFN2 knockout Offspring

defective insulin
signaling
ATP

943 DEGs
slight liver
ER function increased
mitochondrial
phagosome insulinemia
dysfunciton
endocrine resistance
mitophagy

ER
glucose
mitochondrial increased intolerance
dysfunction weight gain
dysregulated
MERCs

Fig. 3  Mitochondrial dynamics in oocytes is essential to fertility DRP1 results in infertility due impaired granulosa cell-oocyte com-
and links mitochondrial-endoplasmic reticulum (ER) dysfunction munication and arrested folliculogenesis. On the other hand, oocytes
with metabolic disorder transmission. Oocyte-specific deletion of lacking mitofusin 2 (MFN2) have a pronounced transcriptomic
dynamin-related protein 1 (DRP1) results in oocytes with elongated change with 943 differentially expressed genes (DEGs) associated
mitochondria, depleted endoplasmic reticulum (ER) calcium ­(Ca2+) with pathways regulating, among others, ER function, the phago-
stores, multiorganelle (i.e., mitochondria, ER, peroxisomes and vesi- some, endocrine resistance, and mitophagy (c). These oocytes also
cles) aggregation, and defective secretory vesicle release (a). In turn, present dysregulated mitochondria-ER contact sites (MERCs) and
mitofusin 1 (MFN1) knockout strongly impairs mitochondrial dynam- mitochondrial dysfunction characterized by mitochondrial aggrega-
ics as evidenced by organelle swelling, clustering, abnormal cristae, tion and swelling, decreased mitochondrial membrane potential,
and inner membrane vesiculation, although mitochondrial shape lower mtDNA copy number, lower nicotinamide adenine dinucleotide
remains unchanged (b). These defects are paralleled by decreased (phosphate) (NAD(P)H) levels, increased FAD levels, and decreased
expression of oxidative phosphorylation subunits, decreased mito- ATP content. Unexpectedly, cristae structure is well developed in
chondrial membrane potential, decreased mitochondrial DNA MFN2-knockout oocytes, better than in wild-type mice. As a result,
(mtDNA) copy number, increased flavin adenine dinucleotide (FAD) oocyte-specific MFN2 deficiency results in offspring with increased
levels, and increased ROS levels. As a result, MFN1-deficient oocytes weight gain and glucose intolerance, abnormalities that associate with
have decreased ATP content, increased apoptosis, and impaired PI3K- slight liver mitochondrial dysfunction, increased insulinemia, and
AKT signaling. Despite these differences, the lack of either MFN1 or impaired insulin signaling

oocytes never ovulated due to a folliculogenesis arrest at depletion (Carvalho et al. 2020; Hou et al. 2019; Zhang
the secondary-to-antral follicular stage, a phenotype that was et al. 2019a). Zhang et al. (2019a) associated these effects
shown to worsen with aging and led to accelerated follicular of MFN1 deletion to accumulation of ceramide in oocytes,

13
Biophysical Reviews

a membrane sphingolipid and an important inducer of apop- 2018; Keleher et al. 2018; Ou et al. 2019; Ruebel et al. 2017;
tosis and cell cycle arrest (Fig. 3b). In turn, Carvalho et al. Sirard, 2019; Wang et al. 2018b), can be passed down the
(2020) found that the additional knockout of MFN2 miti- maternal lineage and contribute with the onset of metabolic
gated the impact of MFN1 deficiency on folliculogenesis, disorders in the following generations.
enabling enhanced mitochondrial health, oocyte growth, and It is currently unclear the precise mechanism by which
ovulation in double-knockout females (Carvalho et al. 2020). obesity affects mitochondria and the ER in oocytes. How-
Moreover, while the single loss of MFN1 altered the expres- ever, it is well known that nutrient abundance can overload
sion of 161 genes, the double loss of MFN1 and MFN2 the ETC with highly charged electrons, resulting in exces-
impacted the expression of 474 genes when compared to sive ROS generation and, eventually, oxidative stress. ROS
wild-type oocytes. Enrichment analysis of transcriptomic can damage mitochondria, leading to mitochondrial frag-
differences linked the effects of single MFN1 deletion to mentation and dysfunction, further aggravating the oxida-
disrupted PI3K-Akt signaling (Fig. 3b), a pathway that tive stress (reviewed by (Kakimoto and Kowaltowski, 2016;
is known to have a fundamental role in oogenesis, being Zorzano et al. 2015)). In this context, MFN2 has been shown
required for granulosa cell-oocyte communication (Dong to exert an important role in adapting the mitochondrial
et al. 1996; Liu et al. 2014; Reddy et al. 2008). In keeping metabolism according to nutrient availability. MFN2 acts
with this, previous reports have shown that MFN2 inhibits through regulation of both mitochondrion-mitochondrion
the PI3K-AKT signaling (Chen et al. 2004, 2014; de Brito and mitochondrion-ER interactions, besides determining
and Scorrano, 2009; Ma et al. 2015b), suggesting that the mitochondrial interaction with other subcellular compart-
phenotype of MFN1-knockout oocytes may be explained, ments such as lipid droplets and lysosomes (reviewed by
at least in part, by a deleterious action of MFN2. Indeed, (Benador et al. 2019; Gordaliza‐Alaguero et al. 2019)).
Mfn1 expression in wild-type oocytes was fivefold higher There is also a bunch of evidence connecting decreased
than that of Mfn2, and the treatment of MFN1-deficient MFN2 expression with metabolic inflexibility, mitochon-
oocytes with an agonist of the PI3K-AKT signaling partially drial dysfunction, ER stress, impaired insulin signaling,
rescued oogenesis/folliculogenesis (Carvalho et al. 2020). leptin resistance, glucose intolerance, obesity, and diabetes
Therefore, mitofusins seem to play distinct, nonredundant (Bach et al. 2003; Mahdaviani et al. 2017; Mourier et al.
roles in oocytes, with MFN1 acting downstream of MFN2 2015; Muñoz et al. 2014; Ngoh et al. 2012; Pich et al. 2005;
to counter its activity. Schneeberger et al. 2013; Sebastian et al. 2012; Sebastián
et al. 2016; Tubbs et al. 2014).
Given the key role of MFN2 in regulating the energetic
Role of mitochondrial dynamics metabolism, Garcia et al. (2020) have investigated whether
in oocyte‑linked transmission of metabolic mouse dams with oocyte-specific deletion of MFN2 deliver
disorders offspring that are prone to metabolic disorders. Earlier
reports have shown that MFN2 is not essential for oocyte
Although environmental and genetic factors are the main development, with little, if any, impact on fertility (Carvalho
determinants of metabolic disorders, female over-nutrition et al. 2020; Hou et al. 2019; Zhang et al. 2019b). In spite
permanently programs the metabolism of offspring (Agar- of this, MFN2-deficient oocytes displayed mitochondrial
wal et al. 2018; Akamine et al. 2010; Jungheim et al. 2010; dysfunction, dysregulated MERCs, and an overt transcrip-
Mdaki et al. 2016; Murrin et al. 2012; Oestreich and Moley tomic change encompassing 943 differentially expressed
2017; Rattanatray et al. 2010; Reynolds et al. 2013; Ruager- genes (Garcia et al. 2020). Enrichment analysis of the tran-
Martin et al. 2010; Shankar et al. 2008; Wyman et al. 2008). scriptomic data revealed an over-representation of genes
Accordingly, maternal obesity has been found to cause lipid related to ER function, phagosome, endocrine resistance,
accumulation in granulosa cells and oocytes, thereby result- and mitophagy (Garcia et al. 2020). These findings are in
ing in oocytes with dysfunctional mitochondria as evidenced keeping with previous reports in which MFN2 was targeted
by their ultrastructural defects, aggregation, lower membrane through either oocyte-specific deletion or RNA interference,
potential, increased ROS, and increased C ­ a2+ (Andreas et al. thereby leading to mitochondrial dysfunction, shortened
2019; Boots et al. 2016; Boudoures et al. 2017; Ferey et al. telomeres, increased apoptosis, and, in turn, poor qual-
2019; Marei et al. 2020; Reynolds et al. 2013; Wu et al. ity oocytes and accelerated follicular depletion (Liu et al.
2010, 2015). In addition, obesity leads to MERC disruption, 2016a; Zhang et al. 2016b, 2019b). Likewise, MFN2 overex-
ER stress, and activation of the unfolded protein response pression, which increased mitochondria-ER juxtaposition in
(UPR) in oocytes (Marei et al. 2019, 2020; Wu et al. 2015; oocytes, led to alterations in the ER such as a disintegrated
Zhao et al. 2017). Thus, besides impacting oocyte health ­ a2+ stores (Wakai et al.
network, clusterization, and loss of C
and fertility, these abnormalities, likely underpinned by 2014). Consequently, offspring derived from MFN2-defi-
abnormal mitochondria and epigenetic marks (Agarwal et al. cient oocytes presented increased weight gain and glucose

13
Biophysical Reviews

intolerance, a phenotype that was linked to decreased insu- Rev Clin Lab Sci 55:71–101.https://​doi.​org/​10.​1080/​10408​363.​
linemia and defective insulin signaling (Garcia et al. 2020). 2017.​14221​09
Akamine EH, Marçal AC, Camporez JP, Hoshida MS, Caperuto LC,
Accordingly, Zhao et al. (2017) have reported that obesity Bevilacqua E, Carvalho CRO (2010) Obesity induced by high-
affects MERCs in oocytes, an effect that is prevented by fat diet promotes insulin resistance in the ovary. J Endocrinol
PACS2 downregulation. Furthermore, Saben et al. (2016) 206:65–74. https://​doi.​org/​10.​1677/​JOE-​09-​0461
have shown that maternal metabolic syndrome leads to Alexander C, Votruba M, Pesch UE, Thiselton DL, Mayeer S,
Moore A, Rodriguez M, Kellner U, Leo-Kottler B, Auburger
persistent transmission of impaired mitochondrial fission G, Bhattacharya SS, Wissinger B (2000) OPA1, encoding a
and decreased OPA1 expression across three generations. dynamin-related GTPase, is mutated in autosomal dominant
Altogether, these findings support a role for mitochondrial optic atrophy linked to chromosome 3q28. Nat Genet 26:211–
dynamics in regulating mother-to-offspring transmission of 215.https://​doi.​org/​10.​1038/​79944
Al-Zubaidi U, Liu J, Cinar O, Robker RL, Adhikari D, Carroll J
metabolic disorders. Further studies should focus on charac- (2019) The spatio-temporal dynamics of mitochondrial mem-
terizing a possible underlying mechanism linking maternal brane potential during oocyte maturation. Mol Hum Reprod
obesity with abnormal mitochondrial dynamics. 25:695–705. https://​doi.​org/​10.​1093/​molehr/​gaz055
Al-Zubaidi U, Adhikari D, Cinar O, Zhang QH, Yuen WS, Murphy
MP, Rombauts L, Robker RL, Carroll J (2021) Mitochondria-
targeted therapeutics, MitoQ and BGP-15, reverse aging-asso-
Concluding remarks ciated meiotic spindle defects in mouse and human oocytes.
Hum Reprod 36:771–784. https://​d oi.​o rg/​1 0.​1 093/​h umrep/​
deaa3​00
A large body of evidence supports that mitochondria are kept Amati-Bonneau P, Valentino ML, Reynier P, Gallardo ME, Born-
under a relatively quiescent state in oocytes, likely to prevent stein B et al (2008) OPA1 mutations induce mitochondrial
the gamete from oxidative stress once somatic granulosa DNA instability and optic atrophy ‘plus’ phenotypes. Brain
cells are responsible for supplying most of the energetic mol- 131:338–351. https://​doi.​org/​10.​1093/​brain/​awm298
Anand R, Wai T, Baker MJ, Kladt N, Schauss AC, Rugarli E, Langer
ecules needed for oogenesis. As such, mitochondrial health T (2014) The i-AAA protease YME1L and OMA1 cleave
is inextricably linked to mitochondrial dynamics, which OPA1 to balance mitochondrial fusion and fission. J Cell Biol
regulates in oocytes several aspects of the mitochondrial 204:919–929. https://​doi.​org/​10.​1083/​jcb.​20130​8006
biology including organelle morphofunctional characteris- Andreas E, Reid M, Zhang W, Moley KH (2019) The effect of mater-
nal high-fat/high-sugar diet on offspring oocytes and early
tics, mobility, interaction with subcellular compartments, embryo development. Mol Hum Reprod 25:717–728. https://​
signal transduction, and apoptosis. Thereby, malfunctioning doi.​org/​10.​1093/​molehr/​gaz049
of mitochondrial dynamics can disturb oocyte formation, Babayev E, Wang T, Szigeti-Buck K, Lowther K, Taylor HS, Horvath
ultimately leading to infertility and disease transmission. T, Seli E (2016) Reproductive aging is associated with changes
in oocyte mitochondrial dynamics, function, and mtDNA quan-
tity. Maturitas 93:121–130.https://​doi.​org/​10.​1016/j.​matur​itas.​
Acknowledgements Marcos R. Chiaratti is funded by the São Paulo 2016.​06.​015
Research Foundation (FAPESP/Brazil – grants # 2017/04372-0 and Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J,
2020/15412-6), Conselho Nacional de Desenvolvimento Científico e Daugaard JR, Lloberas J, Camps M, Zierath JR et al (2003)
Tecnológico (CNPq/Brazil), the Coordenação de Aperfeiçoamento de Mitofusin-2 determines mitochondrial network architecture
Pessoal de Nível Superior (CAPES/Brazil – finance code 001), and the and mitochondrial metabolism. A novel regulatory mechanism
Academy of Medical Sciences-Newton Advanced Fellowship. altered in obesity. J Biol Chem 278:17190–17197. https://​doi.​
org/​10.​1074/​jbc.​M2127​54200
Declarations Baird DT, Collins J, Egozcue J, Evers LH, Gianaroli L, Leridon
H, Sunde A, Templeton A, Van Steirteghem A, Cohen J et al
Ethical approval This article does not contain any studies with human (2005) Fertility and aging. Hum Reprod Update 11:261–276.
participants or animals performed by the author. https://​doi.​org/​10.​1093/​humupd/​dmi006
Ban-Ishihara R, Ishihara T, Sasaki N, Mihara K, Ishihara N (2013)
Dynamics of nucleoid structure regulated by mitochondrial
Conflict of interest The author declares no competing interests. fission contributes to cristae reformation and release of
cytochrome c. Proc Natl Acad Sci U S A 110:11863–11868.
https://​doi.​org/​10.​1073/​pnas.​13019​51110
Benador IY, Veliova M, Liesa M, Shirihai OS (2019) Mitochon-
References dria bound to lipid droplets: where mitochondrial dynamics
regulate lipid storage and utilization. Cell Metab 29:827–835.
Abbassi L, El-Hayek S, Carvalho KF, Wang W, Yang Q, Granados- https://​doi.​org/​10.​1016/j.​cmet.​2019.​02.​011
Aparici S, Mondadori R, Bordignon V, Clarke HJ (2021) Epider- Bentov Y, Yavorska T, Esfandiari N, Jurisicova A, Casper RF (2011)
mal growth factor receptor signaling uncouples germ cells from The contribution of mitochondrial function to reproductive
the somatic follicular compartment at ovulation. Nat Commun aging. J Assist Reprod Genet 28:773–783. https://​doi.​org/​10.​
12:1–13. https://​doi.​org/​10.​1038/​s41467-​021-​21644-z 1007/​s10815-​011-​9588-7
Agarwal P, Morriseau TS, Kereliuk SM, Doucette CA, Wicklow BA, Biggers JD, Whittingham DG, Donahue RP (1967) The pattern of
Dolinsky VW (2018) Maternal obesity, diabetes during preg- energy metabolism in the oocyte and zygote. Proc Natl Acad
nancy and epigenetic mechanisms that influence the develop- Sci U S A 58:560–567. https://​doi.​org/​10.​1073/​pnas.​58.2.​560
mental origins of cardiometabolic disease in the offspring. Crit

13
Biophysical Reviews

Binelli M, Murphy BD (2010) Coordinated regulation of follicle devel- stability in skeletal muscle and tolerance of mtDNA mutations.
opment by germ and somatic cells. Reprod Fertil Dev 22:1–12. Cell 141:280–289. https://​doi.​org/​10.​1016/j.​cell.​2010.​02.​026
https://​doi.​org/​10.​1071/​RD092​18 Chen KH, Dasgupta A, Ding J, Indig FE, Ghosh P, Longo L, D. (2014)
Boots CE, Boudoures A, Zhang W, Drury A, Moley KH (2016) Obe- Role of mitofusin 2 (Mfn2) in controlling cellular proliferation.
sity-induced oocyte mitochondrial defects are partially prevented FASEB J 28:382–394.https://​doi.​org/​10.​1096/​fj.​13-​230037
and rescued by supplementation with co-enzyme Q10 in a mouse Cho YM, Kwon S, Pak YK, Seol HW, Choi YM, Park DJ, Park KS,
model. Hum Reprod 31:2090–2097. https://​doi.​org/​10.​1093/​ Lee HK (2006) Dynamic changes in mitochondrial biogenesis
humrep/​dew181 and antioxidant enzymes during the spontaneous differentiation
Boudoures AL, Saben J, Drury A, Scheaffer S, Modi Z, Zhang W, of human embryonic stem cells. Biochem Biophys Res Commun
Moley KH (2017) Obesity-exposed oocytes accumulate and 348:1472–1478. https://​doi.​org/​10.​1016/j.​bbrc.​2006.​08.​020
transmit damaged mitochondria due to an inability to activate Cipolat S, de Brito OM, Dal Zilio B, Scorrano L (2004) OPA1 requires
mitophagy. Dev Biol 426:126–138. https://​doi.​org/​10.​1016/j.​ mitofusin 1 to promote mitochondrial fusion. Proc Natl Acad
ydbio.​2017.​04.​005 Sci U S A 101:15927–15932. https://d​ oi.o​ rg/1​ 0.1​ 073/p​ nas.0​ 4070​
Brevini TAL, Vassena R, Francisci C, Gandolfi F (2005) Role of adeno- 43101
sine triphosphate, active mitochondria, and microtubules in the Clarke HJ (2017) Regulation of germ cell development by intercellular
acquisition of developmental competence of parthenogenetically signaling in the mammalian ovarian follicle. Wiley Interdiscip
activated pig oocytes. Biol Reprod 72:1218–1223. https://​doi.​ Rev Dev Biol. https://​doi.​org/​10.​1002/​wdev.​294
org/​10.​1095/​biolr​eprod.​104.​038141 Cohen J, Scott R, Schimmel T, Levron J, Willadsen S (1997) Birth of
Brockmann K, Dreha-Kulaczewski S, Dechent P, Bönnemann C, Helms infant after transfer of anucleate donor oocyte cytoplasm into
G et al (2008) Cerebral involvement in axonal Charcot-Marie- recipient eggs. Lancet 350:186–187. https://​doi.​org/​10.​1016/​
Tooth neuropathy caused by mitofusin 2 mutations. J Neurol S0140-​6736(05)​62353-7
255:1049–1058.https://​doi.​org/​10.​1007/​s00415-​008-​0847-1 Craven L, Alston CL, Taylor RW, Turnbull DM (2017) Recent
Cao L, Shitara H, Horii T, Nagao Y, Imai H, Abe K, Hara T, Hayashi Advances in Mitochondrial Disease. Annu Rev Genom-
J-I, Yonekawa H (2007) The mitochondrial bottleneck occurs ics Hum Genet 18:257–275. https://​d oi.​o rg/​1 0.​1 146/​a nnur​
without reduction of mtDNA content in female mouse germ cells. ev-​genom-​091416-​035426
Nat Genet 39:386–390. https://​doi.​org/​10.​1038/​ng1970 Dalton CM, Szabadkai G, Carroll J (2014) Measurement of ATP in
Carelli V, Musumeci O, Caporali L, Zanna C, Morgia CL, Del Dotto single oocytes: Impact of maturation and cumulus cells on levels
V, Porcelli AM, Rugolo M, Valentino ML, Iommarini L et al and consumption. J Cellular Physiol 229:353–361. https://​doi.​
(2015) Syndromic parkinsonism and dementia associated with org/​10.​1002/​jcp.​24457
OPA1 missense mutations. Ann Neurol 78:21–38. https://​doi.​ de Brito OM, Scorrano L (2008) Mitofusin 2 tethers endoplasmic retic-
org/​10.​1002/​ana.​24410 ulum to mitochondria. Nature 456:605–610. https://​doi.​org/​10.​
Carroll J, Swann K, Whittingham D, Whitaker M (1994) Spatiotem- 1038/​natur​e07534
poral dynamics of intracellular [Ca2+](i) oscillations during the de Brito OM, Scorrano L (2009) Mitofusin-2 regulates mitochondrial
growth and meiotic maturation of mouse oocytes. Development and endoplasmic reticulum morphology and tethering: The role
120:3507–3517 of Ras. Mitochondrion 9:222–226. https://d​ oi.o​ rg/1​ 0.1​ 016/j.m ​ ito.​
Carvalho KF, Machado TS, Garcia BM, Zangirolamo AF, Macabelli 2009.​02.​005
CH, Sugiyama FHC, Grejo MP, Augusto Neto JD, Tostes K, Delettre C, Leners G, Friffoin JM, Lorenzo C, Belenguer P, Pelloquin
Ribeiro FKS et al (2020) Mitofusin 1 is required for oocyte L, Grosgeorge J, Turc-Carel C, Perret E, Astarie-Dequeker C,
growth and communication with follicular somatic cells. FASEB Lasquellec L, Arnaud B, Ducommun B, Kaplan J, Hamel CP
J 34:7644–7660. https://​doi.​org/​10.​1096/​fj.​20190​1761R (2000) Nuclear gene OPA1, encoding a mitochondrial dynamin-
Chakrabarti R, Ji W-K, Stan RV, de Juan Sanz J, Ryan TA, Higgs HN related protein, is mutated in dominant optic atrophy. Nat Genet
(2018) INF2-mediated actin polymerization at the ER stimulates 26:207–210. https://​doi.​org/​10.​1038/​79936
mitochondrial calcium uptake, inner membrane constriction, and Diez-Juan A, Rubio C, Marin C, Martinez S, Al-Asmar N, Riboldi
division. J Cell Biol 217:251–268. https://​doi.​org/​10.​1083/​jcb.​ M, Díaz-Gimeno P, Valbuena D, Simón C (2015) Mitochondrial
20170​9111 DNA content as a viability score in human euploid embryos: less
Chen H, Chan DC (2017) Mitochondrial dynamics in regulating the is better. Fertil Steril 104:534-541.e1. https://​doi.​org/​10.​1016/j.​
unique phenotypes of cancer and stem cells. Cell Metab 26:39– fertn​stert.​2015.​05.​022
48. https://​doi.​org/​10.​1016/j.​cmet.​2017.​05.​016 Dong J, Albertini DF, Nishimori K, Kumar TR, Lu N, Matzuk MM
Chen H, Detmer S, a, Ewald, A. J., Griffin, E. E., Fraser, S. E. and (1996) Growth differentiation factor-9 is required during early
Chan, D. C. (2003) Mitofusins Mfn1 and Mfn2 coordinately ovarian folliculogenesis. Nature 383:531–535.https://​doi.​org/​10.​
regulate mitochondrial fusion and are essential for embryonic 1038/​38353​1a0
development. J Cell Biol 160:189–200. https://​doi.​org/​10.​1083/​ Downs SM (1995) The influence of glucose, cumulus cells, and meta-
jcb.​20021​1046 bolic coupling on ATP levels and meiotic control in the isolated
Chen KH, Guo X, Ma D, Guo Y, Li Q, Yang D, Li P, Qiu X, Wen S, mouse oocyte. Dev Biol 167:502–512. https://​doi.​org/​10.​1006/​
Xiao RP et al (2004) Dysregulation of HSG triggers vascular dbio.​1995.​1044
proliferative disorders. Nat Cell Biol 6:872–883. https://​doi.​org/​ Downs SM, Humpherson PG, Leese HJ (2002) Pyruvate utilization by
10.​1038/​ncb11​61 mouse oocytes is influenced by meiotic status and the cumulus
Chen H, Chomyn A, Chan DC (2005) Disruption of fusion results oophorus. Mol Reprod Dev 62:113–123. https://d​ oi.o​ rg/1​ 0.1​ 002/​
in mitochondrial heterogeneity and dysfunction. J Biol Chem mrd.​10067
280:26185–26192. https://​doi.​org/​10.​1074/​jbc.​M5030​62200 Eppig JJ (1976) Analysis of mouse oogenesis in vitro. Oocyte isola-
Chen H, McCaffery JM, Chan DC (2007) Mitochondrial fusion protects tion and the utilization of exogenous energy sources by growing
against neurodegeneration in the cerebellum. Cell 130:548–562. oocytes. J Exp Zool 198:375–381. https://​doi.​org/​10.​1002/​jez.​
https://​doi.​org/​10.​1016/j.​cell.​2007.​06.​026 14019​80311
Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM, Eppig JJ, Pendola FL, Wigglesworth K, Pendola JK (2005) Mouse
Chan DC (2010) Mitochondrial fusion is required for mtDNA oocytes regulate metabolic cooperativity between granulosa

13
Biophysical Reviews

cells and oocytes: Amino acid transport. Biol Reprod 73:351– Hudson G, Amati-Bonneau P, Blakely EL, Stewart JD, He L et al
357. https://​doi.​org/​10.​1095/​biolr​eprod.​105.​041798 (2008) Mutation of OPA1 causes dominant optic atrophy with
Fahrner JA, Liu R, Perry MS, Klein J, Chan DC (2016) A novel de external ophthalmoplegia, ataxia, deafness and multiple mito-
novo dominant negative mutation in DNM1L impairs mito- chondrial DNA deletions: a novel disorder of mtDNA main-
chondrial fission and presents as childhood epileptic encepha- tenance. Brain 131:329–337. https://​d oi.​o rg/​1 0.​1 093/​b rain/​
lopathy. Am J Med Genet A 170:2002–2011. https://​doi.​org/​ awm272
10.​1002/​ajmg.a.​37721 Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda
Fair T, Hulshof SCJ, Hyttel P, Greve T, Boland M (1997) Oocyte K, Otera H, Nakanishi Y, Nonaka I, Goto Y-I et al (2009)
ultrastructure in bovine primordial to early tertiary follicles. Mitochondrial fission factor Drp1 is essential for embryonic
Anat Embryol (berl) 195:327–336. https://​d oi.​o rg/​1 0.​1 007/​ development and synapse formation in mice. Nat Cell Biol
s0042​90050​052 11:958–966. https://​doi.​org/​10.​1038/​ncb19​07
Ferey JLA, Boudoures AL, Reid M, Drury A, Scheaffer S, Modi Z, Ishihara T, Ban-Ishihara R, Maeda M, Matsunaga Y, Ichimura A,
Kovacs A, Pietka T, DeBosch BJ, Thompson MD et al (2019) Kyogoku S, Aoki H, Katada S, Nakada K, Nomura M et al
A maternal high-fat, high-sucrose diet induces transgenera- (2015) Dynamics of mitochondrial DNA nucleoids regulated
tional cardiac mitochondrial dysfunction independently of by mitochondrial fission is essential for maintenance of homo-
maternal mitochondrial inheritance. Am J Physiol Heart Circ geneously active mitochondria during neonatal heart develop-
Physiol 316:H1202–H1210. https://​doi.​org/​10.​1152/​ajphe​art.​ ment. Mol Cell Biol 35:211–223.https://​doi.​org/​10.​1128/​MCB.​
00013.​2019 01054-​14
Fragouli E, Spath K, Alfarawati S, Kaper F, Craig A, Michel C-E, Jansen RPS, De Boer K (1998) The bottleneck: mitochondrial imper-
Kokocinski F, Cohen J, Munne S, Wells D (2015) Altered lev- atives in oogenesis and ovarian follicular fate. Mol Cell Endo-
els of mitochondrial DNA are associated with female age, ane- crinol 145:81–88. https://​d oi.​o rg/​1 0.​1 016/​s 0303-​7 207(98)​
uploidy, and provide an independent measure of embryonic 00173-7
implantation potential. PLoS Genet 11:e1005241. https://​doi.​ Johnson MT, Freeman EA, Gardner DK, Hunt PA (2007) Oxidative
org/​10.​1371/​journ​al.​pgen.​10052​41 metabolism of pyruvate is required for meiotic maturation of
Fyfe JC, Al-Tamimi RA, Liu J, Schaffer AA, Agarwala R, Henthorn PS murine oocytes in vivo1. Biol Reprod 77:2–8. https://​doi.​org/​
(2011) A novel mitofusin 2 mutation causes canine fetal-onset 10.​1095/​biolr​eprod.​106.​059899
neuroaxonal dystrophy. Neurogenetics 12:223–232.https://​doi.​ Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer
org/​10.​1007/​s10048-​011-​0285-6 JE, Moley KH (2010) Diet-induced obesity model: abnormal
Garcia BM, Machado TS, Carvalho KF, Nolasco P, Nociti RP, Del Col- oocytes and persistent growth abnormalities in the offspring.
lado M, Capo Bianco MJD, Grejo MP, Neto JDA, Sugiyama FHC Endocrinol 151:4039–4046
et al (2020) Mice born to females with oocyte-specific deletion Kakimoto PA, Kowaltowski AJ (2016) Effects of high fat diets on
of mitofusin 2 have increased weight gain and impaired glucose rodent liver bioenergetics and oxidative imbalance. Redox Biol
homeostasis. Mol Hum Reprod 26:938–952. https://​doi.​org/​10.​ 8:216–225. https://​doi.​org/​10.​1016/j.​redox.​2016.​01.​009
1093/​molehr/​gaaa0​71 Keleher MR, Zaidi R, Shah S, Oakley ME, Pavlatos C, Idrissi SE,
Gerber S, Charif M, Chevrollier A, Chaumette T, Angebault C, Kane Xing X, Li D, Wang T, Cheverud JM (2018) Maternal high-fat
MS, Paris A, Alban J, Quiles M, Delettre C et al (2017) Muta- diet associated with altered gene expression, DNA methyla-
tions in DNM1L, as in OPA1, result in dominant optic atrophy tion, and obesity risk in mouse offspring. PLoS ONE 13:1–28.
despite opposite effects on mitochondrial fusion and fission. https://​doi.​org/​10.​1371/​journ​al.​pone.​01926​06
Brain 140:2586–2596. https://​doi.​org/​10.​1093/​brain/​awx219 Kleele T, Rey T, Winter J, Zaganelli S, Mahecic D, Perreten Lambert
Giacomello M, Pyakurel A, Glytsou C, Scorrano L (2020) The cell H, Ruberto FP, Nemir M, Wai T, Pedrazzini T et al (2021)
biology of mitochondrial membrane dynamics. Nat Rev Mol Cell Distinct fission signatures predict mitochondrial degradation
Biol 21:204–224. https://​doi.​org/​10.​1038/​s41580-​020-​0210-7 or biogenesis. Nature 593:435–439. https://​doi.​org/​10.​1038/​
Gordaliza-Alaguero I, Cantó C, Zorzano A (2019) Metabolic impli- s41586-​021-​03510-6
cations of organelle–mitochondria communication. EMBO Rep Kobayashi T, Surani MA (2018) On the origin of the human ger-
20:1–27. https://​doi.​org/​10.​15252/​embr.​20194​7928 mline. Development 145:2–5. https://​d oi.​o rg/​1 0.​1 242/​d ev.​
Gustafsson CM, Falkenberg M, Larsson NG (2016) Maintenance 150433
and Expression of Mammalian Mitochondrial DNA. Annu Rev Koch J, Feichtinger RG, Freisinger P, Pies M, Schrödl F, Iuso A,
Biochem 85:133–160. https://​doi.​org/​10.​1146/​annur​ev-​bioch​ Sperl W, Mayr JA, Prokisch H, Haack TB (2016) Disturbed
em-​060815-​014402 mitochondrial and peroxisomal dynamics duet to loss of MFF
Harris SE, Leese HJ, Gosden RG, Picton HM (2009) Pyruvate and causes Leigh-like encephalopathy, optic atrophy and peripheral
oxygen consumption throughout the growth and development of neuropathy. J Med Genet 53:270–278. https://​doi.​org/​10.​1136/​
murine oocytes. Mol Reprod Dev 76:231–238. https://​doi.​org/​ jmedg​enet-​2015-​103500
10.​1002/​mrd.​20945 Korobova F, Ramabhadran V, Higgs HN (2013) An actin-dependent
Hartmann B, Wai T, Hu H, Macvicar T, Musante L, Fischer-Zirnsak B, step in mitochondrial fission mediated by the ER-associated
Stenzel W, Gräf R, van den Heuvel L, Ropers H-H et al (2016) formin INF2. Science 339:464–467. https://​doi.​org/​10.​1126/​
Homozygous YME1L1 mutation causes mitochondriopathy with scien​ce.​12283​60
optic atrophy and mitochondrial network fragmentation. Elife Krasich R, Copeland WC (2017) DNA polymerases in the mitochon-
5:e16078. https://​doi.​org/​10.​7554/​eLife.​16078 dria: a critical review of the evidence. Front Biosci (landmark
Hashimoto S, Morimoto N, Yamanaka M, Matsumoto H, Yamochi T, Ed) 22:692–709. https://​doi.​org/​10.​2741/​4510
Goto H, Inoue M, Nakaoka Y, Shibahara H, Morimoto Y (2017) Krisher RL, Bavister BD (1998) Responses of oocytes and embryos to
Quantitative and qualitative changes of mitochondria in human the culture environment. Theriogenology 49:103–114. https://​
preimplantation embryos. J Assist Reprod Genet 34:573–580. doi.​org/​10.​1016/​s0093-​691x(97)​00405-6
https://​doi.​org/​10.​1007/​s10815-​017-​0886-6 Kristensen SG, Pors SE, Andersen CY (2017) Improving oocyte qual-
Hou X, Zhu S, Zhang H, Li C, Qiu D, Ge J, Guo X, Wang Q (2019) ity by transfer of autologous mitochondria from fully grown
Mitofusin1 in oocyte is essential for female fertility. Redox Biol oocytes. Hum Reprod 32:725–732. https://​doi.​org/​10.​1093/​
21:101110. https://​doi.​org/​10.​1016/j.​redox.​2019.​101110 humrep/​dex043

13
Biophysical Reviews

Kruip TAM, Cran DG, van Beneden TH, Dieleman SJ (1983) Struc- functions in inbred and outbred mice. Sci Rep 10:1–14. https://​
tural changes in bovine oocytes during final maturation in vivo. doi.​org/​10.​1038/​s41598-​020-​66702-6
Gam Res 8:29–47 May-Panloup P, Boucret L, Chao de la Barca J-M, Desquiret-Dumas
Labarta E, de Los Santos MJ, Escribá MJ, Pellicer A, Herraiz S (2019a) V, Ferré-L’Hotellier V, Morinière C, Descamps P, Procaccio V,
Mitochondria as a tool for oocyte rejuvenation. Fertil Steril Reynier P (2016) Ovarian ageing: the role of mitochondria in
111:219–226. https://​doi.​org/​10.​1016/j.​fertn​stert.​2018.​10.​036 oocytes and follicles. Hum Reprod Update 22:725–743. https://​
Labarta E, de Los Santos MJ, Herraiz S, Escribá MJ, Marzal A, Buig- doi.​org/​10.​1093/​humupd/​dmw028
ues A, Pellicer A (2019b) Autologous mitochondrial transfer as Mdaki KS, Larsen TD, Wachal AL, Schimelpfenig MD, Weaver
a complementary technique to intracytoplasmic sperm injection LJ, Dooyema SDR, Louwagie EJ, Baack ML (2016) Maternal
to improve embryo quality in patients undergoing in vitro fertili- high-fat diet impairs cardiac function in offspring of diabetic
zation-a randomized pilot study. Fertil Steril 111:86–96. https://​ pregnancy through metabolic stress and mitochondrial dys-
doi.​org/​10.​1016/j.​fertn​stert.​2018.​09.​023 function. Am J Physiol Heart Circ Physiol 310:H681–H692.
Lewis SC, Uchiyama LF, Nunnari J (2016) ER-mitochondria contacts https://​doi.​org/​10.​1152/​ajphe​art.​00795.​2015
couple mtDNA synthesis with mitochondrial division in human Miller B, Kim SJ, Kumagai H, Mehta HH, Xiang W, Liu J, Yen K,
cells. Science 353:aaf5549. https://d​ oi.o​ rg/1​ 0.1​ 126/s​ cienc​ e.a​ af55​ Cohen P (2020) Peptides derived from small mitochondrial
49 open reading frames: Genomic, biological, and therapeutic
Lieber T, Jeedigunta SP, Palozzi JM, Lehmann R, Hurd TR (2019) implications. Exp Cell Res 393:112056. https://​d oi.​o rg/​1 0.​
Mitochondrial fragmentation drives selective removal of delete- 1016/j.​yexcr.​2020.​112056
rious mtDNA in the germline. Nature 570:380–384. https://​doi.​ Misko AL, Sasaki Y, Tuck E, Milbrandt J, Baloh RH (2012) Mito-
org/​10.​1038/​s41586-​019-​1213-4 fusin2 Mutations Disrupt Axonal Mitochondrial Positioning
Liu K, Zhang H, Risal S, Gorre N, Busayavalasa K, Li X, Shen Y, and Promote Axon Degeneration. J Neurosci 32:4145–4155.
Bosbach B, Brännström M (2014) Somatic cells initiate primor- https://​doi.​org/​10.​1523/​JNEUR​OSCI.​6338-​11.​2012
dial follicle activation and govern the development of dormant Motta PM, Nottola SA, Makabe S, Heyn R (2000) Mitochondrial
oocytes in mice. Curr Biol 24:2501–2508. https://​doi.​org/​10.​ morphology in human fetal and adult female germ cells. Hum
1016/j.​cub.​2014.​09.​023 Reprod 15(Suppl 2):129–147. https://​doi.​org/​10.​1093/​humrep/​
Liu Q, Kang L, Wang L, Zhang L, Xiang W (2016a) Mitofusin 2 regu- 15.​suppl_2.​129
lates the oocytes development and quality by modulating meiosis Mourier A, Motori E, Brandt T, Lagouge M, Atanassov I, Galinier
and mitochondrial function. Sci Rep 6:30561. https://​doi.​org/​10.​ A, Rappl G, Brodesser S, Hultenby K, Dieterich C et al (2015)
1038/​srep3​0561 Mitofusin 2 is required to maintain mitochondrial coenzyme
Liu X-M, Zhang Y-P, Ji S-Y, Li B-T, Tian X, Li D, Tong C, Fan H-Y Q levels. J Cell Biol 208:429–442. https://​doi.​org/​10.​1083/​jcb.​
(2016b) Mitoguardin-1 and -2 promote maturation and the devel- 20141​1100
opmental potential of mouse oocytes by maintaining mitochon- Muñoz JP, Ivanova S, Sánchez-Wandelmer J, Martínez-Cristóbal P,
drial dynamics and functions. Oncotarget 7:1155–1167. https://​ Noguera E, Sancho A, Díaz-Ramos A, Hernández-Alvarez MI,
doi.​org/​10.​18632/​oncot​arget.​6713 Sebastián D, Mauvezin C et al (2014) Mfn2 modulates the UPR
Losón OC, Song Z, Chen H, Chan DC (2013) Fis1, Mff, MiD49, and and mitochondrial function via repression of PERK. EMBO J
MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol 33:171. https://​doi.​org/​10.​1038/​emboj.​2013.​168
Biol Cell 24:659–667. https://d​ oi.o​ rg/1​ 0.1​ 091/m ​ bc.E​ 12-1​ 0-0​ 721 Murrin CM, Kelly GE, Tremblay RE, Kelleher CC (2012) Body mass
Ma H, Folmes CDL, Wu J, Morey R, Mora-Castilla S, Ocampo A, Ma index and height over three generations: Evidence from the Life-
L, Poulton J, Wang X, Ahmed R et al (2015a) Metabolic rescue ways cross-generational cohort study. BMC Public Health 12:81.
in pluripotent cells from patients with mtDNA disease. Nature https://​doi.​org/​10.​1186/​1471-​2458-​12-​81
524:234–238. https://​doi.​org/​10.​1038/​natur​e14546 Ngoh GA, Papanicolaou KN, Walsh K (2012) Loss of mitofusin 2
Ma L, Chang Y, Yu L, He W, Liu Y (2015b) Pro-apoptotic and anti- promotes endoplasmic reticulum stress. J Biol Chem 287:20321–
proliferative effects of mitofusin-2 via PI3K/Akt signaling in 20332. https://​doi.​org/​10.​1074/​jbc.​M112.​359174
breast cancer cells. Oncol Lett 10:3816–3822. https://​doi.​org/​ Oestreich AK, Moley KH (2017) Developmental and Transmittable
10.​3892/​ol.​2015.​3748 Origins of Obesity-Associated Health Disorders. Trends Genet
Magnusson C, Hillensjö T, Hamberger L, Nilsson L (1986) Oxygen 33:399–407. https://​doi.​org/​10.​1016/j.​tig.​2017.​03.​008
consumption by human oocytes and blastocysts grown in vitro. Ota A, Ishihara T, Ishihara N (2020) Mitochondrial nucleoid mor-
Hum Reprod 1:183–184. https://​doi.​org/​10.​1093/​oxfor​djour​nals.​ phology and respiratory function are altered in Drp1-deficient
humrep.​a1363​77 HeLa cells. J Biochem 167:287–294. https://​doi.​org/​10.​1093/​jb/​
Mahdaviani K, Benador IY, Su S, Gharakhanian RA, Stiles L, Trudeau mvz112
KM, Cardamone M, Enríquez-Zarralanga V, Ritou E, Apraha- Ou XH, Zhu CC, Sun SC (2019) Effects of obesity and diabetes on
mian T et al (2017) Mfn2 deletion in brown adipose tissue pro- the epigenetic modification of mammalian gametes. J Cellular
tects from insulin resistance and impairs thermogenesis. EMBO Physiol 234:7847–7855
Rep 18:1123–1138. https://​doi.​org/​10.​15252/​embr.​20164​3827 Palmer CS, Elgass KD, Parton RG, Osellame LD, Stojanovski D, Ryan
Manor, U., Bartholomew, S., Golani, G., Christenson, E., Kozlov, MT (2013) Adaptor proteins MiD49 and MiD51 can act indepen-
M., Higgs, H., Spudich, J. and Lippincott-Schwartz, J. (2015). dently of Mff and Fis1 in Drp1 recruitment and are specific for
A mitochondria-anchored isoform of the actin-nucleating spire mitochondrial fission. J Biol Chem 288:27584–27593. https://​
protein regulates mitochondrial division. eLife 4. https://​doi.​org/​ doi.​org/​10.​1074/​jbc.​M113.​479873
10.​7554/​eLife.​08828 Pasquariello R, Ermisch AF, Silva E, McCormick S, Logsdon D,
Marei WFA, Raemdonck GV, Baggerman G, Bols PEJ, Leroy JLMR Barfield JP, Schoolcraft WB, Krisher RL (2019) Alterations in
(2019) Proteomic changes in oocytes after in vitro maturation in oocyte mitochondrial number and function are related to spindle
lipotoxic conditions are different from those in cumulus cells. Sc defects and occur with maternal aging in mice and humans†.
Rep 9:3673. https://​doi.​org/​10.​1038/​s41598-​019-​40122-7 Biol Reprod 100:971–981. https://​doi.​org/​10.​1093/​biolre/​ioy248
Marei WFA, Smits A, Mohey-Elsaeed O, Pintelon I, Ginneberge Pedersen T, Peters H (1968) Proposal for a classification of oocytes and
D, Bols PEJ, Moerloose K, Leroy JLMR (2020) Differential follicles in the mouse ovary. J Reprod Fertil 17:555–557. https://​
effects of high fat diet-induced obesity on oocyte mitochondrial doi.​org/​10.​1530/​jrf.0.​01705​55

13
Biophysical Reviews

Pernas L, Scorrano L (2016) Mito-morphosis: mitochondrial fusion, three generations maternal metabolic syndrome programs mito-
fission, and cristae remodeling as key mediators of cellular func- chondrial dysfunction via germline changes across three genera-
tion. Annu Rev Physiol 78:505–531. https://​doi.​org/​10.​1146/​ tions. Cell Rep 16:1–8. https://​doi.​org/​10.​1016/j.​celrep.​2016.​05.​
annur​ev-​physi​ol-​021115-​105011 065
Pich S, Bach D, Briones P, Liesa M, Camps M, Testar X, Palacín Saita S, Nolte H, Fiedler KU, Kashkar H, Venne AS, Zahedi RP,
M, Zorzano A (2005) The Charcot-Marie-Tooth type 2A gene Krüger M, Langer T (2017) PARL mediates Smac proteolytic
product, Mfn2, up-regulates fuel oxidation through expression maturation in mitochondria to promote apoptosis. Nat Cell Biol
of OXPHOS system. Hum Mol Genet 14:1405–1415. https://d​ oi.​ 19:318–328. https://​doi.​org/​10.​1038/​ncb34​88
org/​10.​1093/​hmg/​ddi149 Santos TA, El Shourbagy S, St. John, J. C. (2006) Mitochondrial con-
Porterfield DM, Trimarchi JR, Keefe DL, Smith PJ (1998) Characteri- tent reflects oocyte variability and fertilization outcome. Fertil
zation of oxygen and calcium fluxes from early mouse embryos Steril 85:584–591. https://​doi.​org/​10.​1016/j.​fertn​stert.​2005.​09.​
and oocytes. Biol Bull 195:208–209. https://​doi.​org/​10.​2307/​ 017
15428​42 Sawyer SL, Ng AC-H, Innes AM, Wagner JD, Dyment DA et al (2015)
Pozzi A, Dowling DK, Sloan D (2019) The genomic origins of small Homozygous mutations in MFN2 cause multiple symmetric lipo-
mitochondrial RNAs: are they transcribed by the mitochon- matosis associated with neuropathy. Hum Mol Genet 24:5109–
drial dna or by mitochondrial pseudogenes within the nucleus 5114. https://​doi.​org/​10.​1093/​hmg/​ddv229
(NUMTs)? Genome Biol Evol 11:1883–1896. https://d​ oi.o​ rg/1​ 0.​ Schaaf CP, Blazo M, Lewis RA, Tonini RE, Takei H, Wang J, Wong
1093/​gbe/​evz132 L-J, Scaglia F (2011) Early-onset severe neuromuscular pheno-
Prigione A, Rohwer N, Hoffmann S, Mlody B, Drews K, Bukowiecki type associated with compound heterozygosity foor OPA1 muta-
R, Blümlein K, Wanker EE, Ralser M, Cramer T et al (2014) tions. Mol Genet Metab 103:383–387. https://​doi.​org/​10.​1016/j.​
HIF1 alpha modulates cell fate reprogramming through early ymgme.​2011.​04.​018
glycolytic shift and upregulation of PDK1-3 and PKM2. Stem Schneeberger M, Dietrich MO, Sebastián D, Imbernón M, Castaño
Cells 32:364–376. https://​doi.​org/​10.​1002/​stem.​1552 C, Garcia A, Esteban Y, Gonzalez-Franquesa A, Rodríguez IC,
Rath, S., Sharma, R., Gupta, R., Ast, T., Chan, C., Durham, T. J., Good- Bortolozzi A et al (2013) Mitofusin 2 in POMC neurons con-
man, R. P., Grabarek, Z., Haas, M. E., Hung, W. H. W., et al. nects ER stress with leptin resistance and energy imbalance. Cell
(2020). MitoCarta3.0: an updated mitochondrial proteome now 155:172–187. https://​doi.​org/​10.​1016/j.​cell.​2013.​09.​003
with sub-organelle localization and pathway annotations. Nucleic Schwartz D, Mayaux MJ (1982) Female fecundity as a function of
Acids Res 1–7. https://​doi.​org/​10.​1093/​nar/​gkaa1​011 age: results of artificial insemination in 2193 nulliparous women
Rattanatray L, MacLaughlin SM, Kleemann DO, Walker SK, Muh- with azoospermic husbands. Federation CECOS New Engl J Med
lhausler BS, McMillen IC (2010) Impact of maternal pericon- 306:404–406. https://​doi.​org/​10.​1056/​NEJM1​98202​18306​0706
ceptional overnutrition on fat mass and expression of adipogenic Sebastian D, Hernandez-Alvarez MI, Segales J, Sorianello E, Munoz
and lipogenic genes in visceral and subcutaneous fat depots in JP, Sala D, Waget A, Liesa M, Paz JC, Gopalacharyulu P et al
the postnatal lamb. Endocrinology 151:5195–5205. https://​doi.​ (2012) Mitofusin 2 (Mfn2) links mitochondrial and endoplasmic
org/​10.​1210/​en.​2010-​0501 reticulum function with insulin signaling and is essential for nor-
Reddy P, Liu L, Adhikari D, Jagarlamudi K, Rajareddy S, Shen Y, Du mal glucose homeostasis. Proc Natl Acad Sci U S A 109:5523–
C, Tang W, Hämäläinen T, Peng SL et al (2008) Oocyte-specific 5528. https://​doi.​org/​10.​1073/​pnas.​11082​20109
deletion of Pten causes premature activation of the primordial Sebastián D, Sorianello E, Segalés J, Irazoki A, Ruiz-Bonilla V, Sala
follicle pool. Science 319:611–613. https://d​ oi.o​ rg/1​ 0.1​ 126/s​ cien​ D, Planet E, Berenguer-Llergo A, Muñoz JP, Sánchez-Feutrie
ce.​11522​57 M et al (2016) Mfn2 deficiency links age-related sarcopenia
Reynolds RM, Allan KM, Raja EA, Bhattacharya S, McNeill G, Han- and impaired autophagy to activation of an adaptive mitophagy
naford PC, Sarwar N, Lee AJ, Bhattacharya S, Norman JE (2013) pathway. EMBO J 35:e201593084. https://​doi.​org/​10.​15252/​
Maternal obesity during pregnancy and premature mortality from embj.​20159​3084
cardiovascular event in adult offspring: follow-up of 1 323 275 Seli E (2016) Mitochondrial DNA as a biomarker for in-vitro fertiliza-
person years. BMJ 347:f4539. https://d​ oi.o​ rg/1​ 0.1​ 136/b​ mj.f​ 4539 tion outcome. Curr Opin Obstet Gynecol 28:158–163. https://​
Richani D, Dunning KR, Thompson JG, Gilchrist RB (2021) Metabolic doi.​org/​10.​1097/​GCO.​00000​00000​000274
co-dependence of the oocyte and cumulus cells: essential role Shamseldin HE, Alshammari M, Al-Sheddi T, Salih MA, Alkhalidi
in determining oocyte developmental competence. Hum Reprod H, Kentab A, Repetto GM, Hashem M, Alkuraya FS (2012)
Update 27:27–47. https://​doi.​org/​10.​1093/​humupd/​dmaa0​43 Genomic analysis of mitochondrial diseases in a consanguineous
Rimon-Dahari N, Yerushalmi-Heinemann L, Alyagor L, Dekel N population reveals novel candidate disease genes. J Med Genet
(2016) Ovarian folliculogenesis. Results Probl Cell Differ 49:234–241. https://​doi.​org/​10.​1136/​jmedg​enet-​2012-​100836
58:167–190. https://​doi.​org/​10.​1007/​978-3-​319-​31973-5_7 Shankar K, Harrell A, Liu X, Gilchrist JM, Ronis MJJ, Badger TM
Rouzier C, Bannwarth S, Chaussenot A, Chevrollier A, Verschueren (2008) Maternal obesity at conception programs obesity in the
A et al (2012) The MFN2 gene is responsible for mitochon- offspring. Am J Physiol Regul Integr Comp Physiol 294:528–
drial DNA instability and optic atrophy ‘plus’ phenotype. Brain 538. https://doi.org/10.1152/ajpregu.00316.2007
135:23–34. https://​doi.​org/​10.​1093/​brain/​awr323 Sheffer R, Douiev L, Edvardson S, Avraham S, Tamimi K, Soiferman
Ruager-Martin R, Hyde MJ, Modi N (2010) Maternal obesity and D, Meiner V, Saada A (2016) Postnatal microcephaly and pain
infant outcomes. Early Hum Dev 86:715–722. https://​doi.​org/​ insensitivity due to a de novo heterozygous DNM1L mutation
10.​1016/j.​earlh​umdev.​2010.​08.​007 causing impaired mitochondrial fission and function. Am J Med
Ruebel ML, Cotter M, Sims CR, Moutos DM, Badger TM, Cleves MA, Genet A 170:1603–1607. https://​doi.​org/​10.​1002/​ajmg.a.​37624
Shankar K, Andres A (2017) Obesity modulates inflammation Shen Q, Yamano K, Head BP, Kawajiri S, Cheung JT et al (2014)
and lipid metabolism oocyte gene expression: a single-cell tran- Mutations in Fis1 disrupt orderly disposal of defective mito-
scriptome perspective. J Clin Endocrinol Metab 102:2029–2038. chondria. Mol Biol Cell 25:145–159. https://​doi.​org/​10.​1091/​
https://​doi.​org/​10.​1210/​jc.​2016-​3524 mbc.​E13-​09-​0525
Saben JL, Boudoures AL, Asghar Z, Cusumano A, Scheaffer S, Moley Sirard M-A (2019) Distribution and dynamics of mitochondrial DNA
KH, Saben JL, Boudoures AL, Asghar Z, Thompson A et al methylation in oocytes, embryos and granulosa cells. Sci Rep
(2016) Mitochondrial dysfunction via germline changes across 9:11937. https://​doi.​org/​10.​1038/​s41598-​019-​48422-8

13
Biophysical Reviews

Son MJ, Kwon Y, Son M-Y, Seol B, Choi H-S, Ryu S-W, Choi C, selective fusion govern mitochondrial segregation and elimina-
Cho YS (2015) Mitofusins deficiency elicits mitochondrial meta- tion by autophagy. EMBO J 27:433–46. https://​doi.​org/​10.​1038/​
bolic reprogramming to pluripotency. Cell Death Differ 22:1–13. sj.​emboj.​76019​63
https://​doi.​org/​10.​1038/​cdd.​2015.​43 Udagawa O, Ishihara T, Maeda M, Matsunaga Y, Tsukamoto S,
Spiegel R, Saada A, Flannery PJ, Burté F, Soiferman D, Khayat M, Kawano N, Miyado K, Shitara H, Yokota S, Nomura M et al
Eisner V, Vladovski R, Taylor RW, Bindoff LA et al (2016) Fatal (2014) Mitochondrial fission factor Drp1 maintains oocyte qual-
infantile mitochonddrial encephalomyopathy, hypertrophic car- ity via dynamic rearrangement of multiple organelles. Curr Biol
diomyopathy and optic artrophy associated with a homozygous 24:2451–2458. https://​doi.​org/​10.​1016/j.​cub.​2014.​08.​060
OPA1 mutation. J Med Genet 53:127–131. https://​doi.​org/​10.​ Van Blerkom J, Davis PW, Lee J (1995) ATP content of human oocytes
1136/​jmedg​enet-​2015-​103361 and developmental potential and outcome after in-vitro fertiliza-
Spinelli JB, Haigis MC (2018) The multifaceted contributions of tion and embryo transfer. Hum Reprod 10:415–424. https://​doi.​
mitochondria to cellular metabolism. Nat Cell Biol 20:745–754. org/​10.​1093/​oxfor​djour​nals.​humrep.​a1359​54
https://​doi.​org/​10.​1038/​s41556-​018-​0124-1 Van Blerkom J, Davis P, Alexander S (2000) Differential mitochondrial
Stewart JB, Chinnery PF (2020) Extreme heterogeneity of human distribution in human pronuclear embryos leads to dispropor-
mitochondrial DNA from organelles to populations. Nature Rev tionate inheritance between blastomeres: relationship to micro-
Genet 22:106–118. https://d​ oi.o​ rg/1​ 0.1​ 038/s​ 41576-0​ 20-0​ 0284-x tubular organization, ATP content and competence. Hum Reprod
Stojkovic M, Machado SA, Stojkovic P, Zakhartchenko V, Hutzler P, 15:2621–2633. https://​doi.​org/​10.​1093/​humrep/​15.​12.​2621
Gonçalves PB, Wolf E (2001) Mitochondrial distribution and Van Noord-Zaadstra BM, Looman CWN, Alsbach H, Habbema JDF,
adenosine triphosphate content of bovine oocytes before and Te Velde ER, Karbaat J (1991) Delaying childbearing: effect of
after in vitro maturation: correlation with morphological crite- age on fecundity and outcome of pregnancy. Bmjl 302:1361–
ria and developmental capacity after in vitro fertilization and 1365. https://​doi.​org/​10.​1136/​bmj.​302.​6789.​1361
culture. Biol Reprod 64:904–909. https://​doi.​org/​10.​1095/​biolr​ Vanstone JR, Smith AM, McBride S, Naas T, Holcik M et al (2016)
eprod​64.3.​904 DNM1L-related mitochondrial fission defect presenting as
Su Y-Q, Sugiura K, Wigglesworth K, O’Brien MJ, Affourtit JP, Pangas refractory epilepsy. Eur J Hum Genet 24:1084–1088. https://​
SA, Matzuk MM, Eppig JJ (2007) Oocyte regulation of meta- doi.​org/​10.​1038/​ejhg.​2015.​243
bolic cooperativity between mouse cumulus cells and oocytes: Von Spiczak S, Helbig KL, Shinde DN, Huether R, Pendziwiat M
BMP15 and GDF9 control cholesterol biosynthesis in cumulus et al (2017) DNM1 encephalopathy: a new disease of vesicle
cells. Development 135:111–121. https://​doi.​org/​10.​1242/​dev.​ fission. Neurology 89:385–394. https://​doi.​org/​10.​1212/​WNL.​
009068 00000​00000​004152
Su YQ, Sugiura K, Eppig JJ (2009) Mouse oocyte control of granulosa Wai T, Teoli D, Shoubridge EA (2008) The mitochondrial DNA
cell development and function: Paracrine regulation of cumulus genetic bottleneck results from replication of a subpopulation
cell metabolism. Semin Reprod Med 27:32–42. https://​doi.​org/​ of genomes. Nat Genet 40:1484–1488. https://​doi.​org/​10.​1038/​
10.​1055/s-​0028-​11080​08 ng.​258
Sugiura K, Su Y-Q, Diaz FJ, Pangas SA, Sharma S, Wigglesworth K, Wai T, Ao A, Zhang X, Cyr D, Dufort D, Shoubridge EA (2010) The
O’Brien MJ, Matzuk MM, Shimasaki S, Eppig JJ (2007) Oocyte- role of mitochondrial DNA copy number in mammalian fertil-
derived BMP15 and FGFs cooperate to promote glycolysis in ity. Biol Reprod 83:52–62. https://​doi.​org/​10.​1095/​biolr​eprod.​
cumulus cells. Development 134:2593–2603. https://​doi.​org/​10.​ 109.​080887
1242/​dev.​006882 Wakai T, Harada Y, Miyado K, Kono T (2014) Mitochondrial dynam-
Tarazona AM, Rodríguez JI, Restrepo LF, Olivera-Angel M (2006) ics controlled by mitofusins define organelle positioning and
Mitochondrial activity, distribution and segregation in bovine movement during mouse oocyte maturation. Mol Hum Reprod
oocytes and in embryos produced in vitro. Reprod Domest Anim 20:1090–1100. https://​doi.​org/​10.​1093/​molehr/​gau064
41:5–11. https://​doi.​org/​10.​1111/j.​1439-​0531.​2006.​00615.x Wang T, Babayev E, Jiang Z, Li G, Zhang M, Esencan E, Horvath T,
Thompson JG, Partridge RJ, Houghton FD, Cox CI, Leese HJ (1996) Seli E (2018a) Mitochondrial unfolded protein response gene
Oxygen uptake and carbohydrate metabolism by in vitro derived Clpp is required to maintain ovarian follicular reserve during
bovine embryos. J Reprod Fertil 106:299–306. https://d​ oi.o​ rg/1​ 0.​ aging, for oocyte competence, and development of pre-implan-
1530/​jrf.0.​10602​99 tation embryos. Aging Cell 17:1–13. https://​doi.​org/​10.​1111/​
Tilly JL, Sinclair DA (2013) Germline energetics, aging, and female acel.​12784
infertility. Cell Metab 17:838–850. https://​doi.​org/​10.​1016/j.​ Wang Q, Tang S-B, Song X-B, Deng T-F, Zhang T-T, Yin S, Luo S-M,
cmet.​2013.​05.​007 Shen W, Zhang C-L, Ge Z-J (2018b) High-glucose concentra-
Trebichalská Z, Kyjovská D, Kloudová S, Otevřel P, Hampl A, Hol- tions change DNA methylation levels in human IVM oocytes.
ubcová Z (2021) Cytoplasmic maturation in human oocytes: An Hum Reprod 33:474–481. https://​d oi.​o rg/​1 0.​1 093/​h umrep/​
ultrastructural study. Biol Reprod 104:106–116. https://​doi.​org/​ dey006
10.​1093/​biolre/​ioaa1​74 Wassarman PM, Josefowicz WJ (1978) Oocyte development in the
Trimarchi JR, Liu L, Porterfield DM, Smith PJ, Keefe DL (2000) mouse: An ultrastructural comparison of oocytes isolated at
Oxidative phosphorylation-dependent and -independent oxygen various stages of growth and meiotic competence. J Morphol
consumption by individual preimplantation mouse embryos. Biol 156:209–235. https://​doi.​org/​10.​1002/​jmor.​10515​60206
Reprod 62:1866–1874. https://​doi.​org/​10.​1095/​biolr​eprod​62.6.​ Waterham HR, Koster J, van Roermund CW, Mooyer PA, Wanders RJ,
1866 Leonard JV (2007) A lethal defect of mitochondrial and peroxi-
Tubbs E, Theurey P, Vial G, Bendridi N, Bravard A, Chauvin MA, somal fission. N Engl J Med 356:1736–1741. https://​doi.​org/​10.​
Ji-Cao J, Zoulim F, Bartosch B, Ovize M et al (2014) Mito- 1056/​NEJMo​a0644​36
chondria-associated endoplasmic reticulum membrane (MAM) Woods DC, Tilly JL (2015) Autologous germline mitochondrial energy
integrity is required for insulin signaling and is implicated in transfer (AUGMENT) in human assisted reproduction. Semin
hepatic insulin resistance. Diabetes 63:3279–3294. https://​doi.​ Reprod Med 33:410–421. https://​doi.​org/​10.​1055/s-​0035-​15678​
org/​10.​2337/​db13-​1751 26
Twig G, Elorza A, Molina AJA, Mohamed H, Wikstrom JD, Walzer Wright VC, Chang J, Jeng G, Macaluso M, Centers for Disease Control
G, Stiles L, Haigh SE, Katz S, Las G et al (2008) Fission and and Prevention (CDC) (2008) Assisted reproductive technology

13
Biophysical Reviews

surveillance–United States, 2005. MMWR Surveill Summ Zhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott R III, Horvath
57:1–23 T, Seli E (2019a) Mitofusin 1 is required for female fertility and
Wu LLY, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ, to maintain ovarian follicular reserve. Cell Death Dis 10:560.
Robker RL (2010) High-fat diet causes lipotoxicity responses in https://​doi.​org/​10.​1038/​s41419-​019-​1799-3
cumulus - Oocyte complexes and decreased fertilization rates. Zhang M, Bener MB, Jiang Z, Wang T, Esencan E, Scott R, Horvath
Endocrinology 151:5438–5445. https://​doi.​org/​10.​1210/​en.​ T, Seli E (2019b) Mitofusin 2 plays a role in oocyte and folli-
2010-​0551 cle development, and is required to maintain ovarian follicular
Wu LL, Russell DL, Wong SL, Chen M, Tsai T-S, St John JC, Norman reserve during reproductive aging. Aging 11:3919–3938. https://​
RJ, Febbraio MA, Carroll J, Robker RL (2015) Mitochondrial doi.​org/​10.​18632/​aging.​102024
dysfunction in oocytes of obese mothers: transmission to off- Zhao L, Lu T, Gao L, Fu X, Zhu S, Hou Y (2017) Enriched endoplas-
spring and reversal by pharmacological endoplasmic reticulum mic reticulum-mitochondria interactions result in mitochondrial
stress inhibitors. Development 142:681–691. https://​doi.​org/​10.​ dysfunction and apoptosis in oocytes from obese mice. J A Sci
1242/​dev.​114850 Biotechnol 8:62. https://​doi.​org/​10.​1186/​s40104-​017-​0195-z
Wyman A, Pinto AB, Sheridan R, Moley KH (2008) One-cell zygote Zhu D, Kennerson ML, Walizada G, Züchner S, Vance JM, Nicholson
transfer from diabetic to nondiabetic mouse results in congenital GA (2005) Charcot–Marie–Tooth with pyramidal signs is geneti-
malformations and growth retardation in offspring. Endocrinol- cally heterogeneous: families with and without MFN2 mutations.
ogy 149:466–469. https://​doi.​org/​10.​1210/​en.​2007-​1273 Neurology 65:496–497. https://​doi.​org/​10.​1212/​01.​wnl.​00001​
Yu Y, Dumollard R, Rossbach A, Lai FA, Swann K (2010) Redistribu- 71345.​62270.​29
tion of mitochondria leads to bursts of ATP production during Zorzano A, Hernández-Alvarez MI, Sebastián D, Muñoz JP (2015)
spontaneous mouse oocyte maturation. J Cell Physiol 224:672– Mitofusin 2 as a driver that controls energy metabolism and insu-
680. https://​doi.​org/​10.​1002/​jcp.​22171 lin signaling. Antioxid Redox Signal 22:1020–1031. https://​doi.​
Zhang H, Liu K (2015) Cellular and molecular regulation of the activa- org/​10.​1089/​ars.​2014.​6208
tion of mammalian primordial follicles: somatic cells initiate fol- Züchner S, Mersiyanova IV, Muglia M, Bissar-Tadmouri N, Rochelle
licle activation in adulthood. Hum Reprod Update 21:779–786. J, Dadali EL, Zappia M, Nelis E, Patitucci A, Senderek J et al
https://​doi.​org/​10.​1093/​humupd/​dmv037 (2004) Mutations in the mitochondrial GTPase mitofusin 2 cause
Zhang Y, Liu X, Bai J, Tian X, Zhao X, Liu W, Duan X, Shang W, Fan Charcot-Marie-Tooth neuropathy type 2A. Nat Genet 36:449–
HY, Tong C (2016a) Mitoguardin regulates mitochondrial fusion 451. https://​doi.​org/​10.​1038/​ng1341
through mitopld and is required for neuronal homeostasis. Mol
Cell 61:111–124. https://​doi.​org/​10.​1016/j.​molcel.​2015.​11.​017 Publisher's note Springer Nature remains neutral with regard to
Zhang J-H, Zhang T, Gao S-H, Wang K, Yang X-Y, Mo F-F, Na Yu, jurisdictional claims in published maps and institutional affiliations.
An T, Li Y-F, Hu J-W et al (2016b) Mitofusin-2 is required for
mouse oocyte meiotic maturation. Sci Rep 6:30970. https://​doi.​
org/​10.​1038/​srep3​0970

13

You might also like