Fcell 12 1347286

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

TYPE Review

PUBLISHED 23 February 2024


DOI 10.3389/fcell.2024.1347286

Effects of reactive oxygen species


OPEN ACCESS and mitochondrial dysfunction on
EDITED BY
Igor Golic,
University of Belgrade, Serbia
reproductive aging
REVIEWED BY
Sarah Scalercio, Jiangbo Song 1†, Li Xiao 1†, Zhehao Zhang 1, Yujin Wang 1,
Instituto de Ciência e Tecnologia em
Biomodelos (ICTB), Brazil
Panayiotis Kouis 2, Lene Juel Rasmussen 2 and Fangyin Dai 1*
Kulvinder Kochar Kaur, 1
State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding,
Kulvinder Kaur Centre For Human Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest
Reproduction, India University, Chongqing, China, 2Center for Healthy Aging, Department of Cellular and Molecular
*CORRESPONDENCE Medicine, University of Copenhagen, Copenhagen, Denmark
Fangyin Dai,
[email protected]

These authors have contributed equally to
this work Mitochondria, the versatile organelles crucial for cellular and organismal viability,
RECEIVED 19 December 2023 play a pivotal role in meeting the energy requirements of cells through the
ACCEPTED 12 February 2024 respiratory chain located in the inner mitochondrial membrane, concomitant
PUBLISHED 23 February 2024
with the generation of reactive oxygen species (ROS). A wealth of evidence
CITATION
derived from contemporary investigations on reproductive longevity strongly
Song J, Xiao L, Zhang Z, Wang Y, Kouis P,
Rasmussen LJ and Dai F (2024), Effects of indicates that the aberrant elevation of ROS level constitutes a fundamental factor
reactive oxygen species and mitochondrial in hastening the aging process of reproductive systems which are responsible for
dysfunction on reproductive aging. transmission of DNA to future generations. Constant changes in redox status,
Front. Cell Dev. Biol. 12:1347286.
doi: 10.3389/fcell.2024.1347286 with a pro-oxidant shift mainly through the mitochondrial generation of ROS, are
linked to the modulation of physiological and pathological pathways in gametes
COPYRIGHT
© 2024 Song, Xiao, Zhang, Wang, Kouis, and reproductive tissues. Furthermore, the quantity and quality of mitochondria
Rasmussen and Dai. This is an open-access essential to capacitation and fertilization are increasingly associated with
article distributed under the terms of the
reproductive aging. The article aims to provide current understanding of the
Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other contributions of ROS derived from mitochondrial respiration to the process of
forums is permitted, provided the original reproductive aging. Moreover, understanding the impact of mitochondrial
author(s) and the copyright owner(s) are
dysfunction on both female and male fertility is conducive to finding
credited and that the original publication in this
journal is cited, in accordance with accepted therapeutic strategies to slow, prevent or reverse the process of gamete
academic practice. No use, distribution or aging, and thereby increase reproductive longevity.
reproduction is permitted which does not
comply with these terms.
KEYWORDS

aging, mitochondria, ROS, gamete quality, fertility, reproductive health

1 Introduction
Throughout history, aging has been a major concern for society. Healthcare advances
and the distribution of vaccines have contributed to the increase in life expectancy from
35 years in the 18th century to 72.6 years today (Nakamura et al., 2017; Mansouri
Torghabeh et al., 2022). Based on numerous observations, it has been hypothesized that
the poor quality of reproductive cells can result in accelerated aging and a shorter

Abbreviations: CAT, catalase; CM, cristae membrane; Drp1, dynamin related protein 1; FADH2, flavin
adenine nucleotide phosphate; FUNDC1, FUN14 domain containing1; GC, granulosa cell; GPX,
glutathione peroxidase; GSR, glutathione reductase; IBM, inner boundary membrane; ICS, intracristal
space; IMM, inner membrane; IMS, intermembrane space; Mfn1/2, mitofuscin 1/2; mtDNA,
mitochondrial DNA; NADH, nicotinamide adenine dinucleotide hydride; OMM, outer membrane; OS,
oxidative stress; OxPhos, oxidative phosphorylation; PRDX, peroxiredoxin; ROS, reactive oxygen species;
SOD, superoxide.

Frontiers in Cell and Developmental Biology 01 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

healthspan (Loose et al., 2022). The state of the reproductive system and the integrity of mitochondrial DNA (mtDNA) also play
is not only essential for fertility, but also for overall health. significant roles (Terman et al., 2007; Noh et al., 2023). Delayed
Reproductive aging is a universal developmental process blastocyst development and reduced quality in gametes are
conserved across species coinciding with age-related fertility associated with accumulated oxidative damage and inefficient
depletion and decline of gamete quality, culminating in infertility clearance of dysfunctional mitochondria (Shen et al., 2021; Khan
and deleterious consequences on the offspring (Drori and Folman, et al., 2023). Importantly, there is a high probability that these issues
1976; Drewry et al., 2011; Archie et al., 2014; Sinha and Rae, 2014). are interconnected.
Reproduction is an energy-costly undertaking that profoundly Here, we provide an overview of the evidence that explain how
impacts on multiple individual characteristics at molecular, mitochondrial function and OS can affect reproductive function and
cellular, and endocrine levels (Loose et al., 2022; Secomandi the rate at which it undergoes cellular age-related alterations.
et al., 2022). Nevertheless, reproductive fitness also offers notable
physiological benefits. For instance, it is commonly discovered that
women who remain reproductively healthy until their late 30 years 2 Mitochondria and ROS
or even 40 years have longer lifespans compared to younger women
who give birth in their late 20 years (Murphy, 2023). Similarly, 2.1 The structure and functions of
research on humans indicates that early menopause may be mitochondria
associated with increased mortality (Shuster et al., 2010). Despite
the presence of numerous intrinsic and extrinsic factors that may Mitochondria are ubiquitous intracellular organelles present in
contribute to aging traits in an organism’s germline, the intricate nearly all eukaryotes and involved in multiple aspects of cellular life,
cellular mechanisms underlying reproductive aging remain poorly with a primary role in energy production. This functional diversity is
understood and warrant further investigation to fully comprehend reflected in the complicated mitochondrial ultrastructure (Figure 1).
the complex interactions involved. Mitochondria, unlike most organelles, are highlighted by their
In recent academic research, there is a growing emphasis on the possession of two distinct membranes: an outer membrane
role of mitochondria as a central component in cellular events (OMM) that encloses the organelle and interfaces with the
associated with reproductive aging primarily due to oxidative cytosol, and an inner membrane (IMM) that displays
stress (OS) caused by the progressive accumulation of ROS, as a morphological complexity. The IMM can be further subdivided
result of oxidative phosphorylation (OxPhos) (Gumienny et al., into two components: the inner boundary membrane (IBM), which
1999; Terman et al., 2007; Roger et al., 2017). Nevertheless, OS is not is closely attached to the outer membrane, and the cristae membrane
the sole contributing factor to mitochondria-dependent changes in (CM), which forms lamellar or tubular protrusion within the
reproductive aging; energy metabolism, mitochondrial dynamics, organelle. These membranes give rise to distinct compartments

FIGURE 1
Mitochondrial ultrastructure and the vicious loop among extensive ROS, mito-structural damage, and mitochondrial dysfunction (partially refer to
Iovine et al., 2021). Mitochondrion is precisely compartmentalized, which can be divided into mtDNA, membrane compartments and aqueous
compartments. The membrane compartments include OMM, IBM, and CM from the outside in. The aqueous parts contain IMS, ICS, and the innermost
matrix. Respiratory chain present within the IBM is composed of CI to V, which undergoes OxPhos for material and energy metabolism accompanied
by ROS production. Once ROS is produced extensively and accumulated constantly in cells, ROS would damage the vulnerable mtDNA and
mitochondrial inner structure, leading to mitochondrial dysfunction and more ROS. mtDNA, mitochondrial DNA; OMM, outer membrane; IBM, inner
boundary membrane; CM, cristae membrane; IMS, intermembrane space; ICS, intracristal space; CI to V, Complex I to V; AQP, aquaporin; VDAC, voltage-
dependent anion channel.

Frontiers in Cell and Developmental Biology 02 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

within the mitochondria, including the intermembrane space (IMS) energy derived from the oxidation of hydrogen carriers is effectively
located between the OMM and the IBM, the intracristal space (ICS) conserved in the form of ATP (Figure 1).
enclosed by the CM, and the innermost matrix (Iovine et al., 2021). From the Free radical theory of aging proposed by Harman early
Particularly, mitochondria possess their own DNA, which encodes in 1956 focusing to the endogenous free radicals to the polished
numerous vital proteins necessary for the assembly and operation of Mitochondrial theory of aging developed to include all forms of ROS
mitochondrial respiratory complexes. MtDNA is especially today, intense research has approved that cumulative oxidative
vulnerable to stress-induced damage believed to be a result of the damage contributes to physiological deterioration and ultimately
absence of histones in structure and limited effectiveness of repair leads to aging and death, and mitochondria play an equally
mechanisms (Kim et al., 2015; Vadakedath et al., 2023). important role (Harman, 1956; Lee and Wei, 2001; Salmon et al.,
Mitochondria are involved in essential functions of the cell, 2010; Holmstrom and Finkel, 2014). Concurrent alterations in ROS
including energy metabolism, substance metabolism, signal level and mitochondrial morphology have been reported in
transduction, intracellular calcium homeostasis, cell apoptosis numerous experimental studies. For example, muscle- and skin-
(Iovine et al., 2021). Energy metabolism and material metabolism derived fibroblasts with an inactive Complex I exhibit an elevated
within mitochondria are synchronized. Mitochondria produce high- ROS level and progressive mitochondrial fragmentation
energy phosphorylated substance ATP into all cellular characterized by reduced mitochondrial length and branch,
compartments through a complex interconnected metabolic whereas patient cells without Complex I deficiency display only
network (TCA cycle, OxPhos, and fatty acid β-oxidation) using controllable ROS levels and a normal mitochondrial morphology
multiple energy sources, such as amino acids, lipids, and (Koopman et al., 2007; Blanchet et al., 2011). The excessive presence
carbohydrate derivatives (Heldt, 1972; Wang J. et al., 2009). of ROS heightens the likelihood of mutations of mtDNA that
Mitochondria possess the ability to modulate energy production encodes proteins related to the respiratory chain, ultimately
via glycolysis, the TCA cycle, and OxPhos in response to alterations affecting ATP production and potent kinetic energy supply for
in cellular energy demands (Lim et al., 2011). Furthermore, motion (Venkatesh et al., 2009). Collectively, mitochondria as the
mitochondria assume a pivotal role in the regulation of primary generators and the recipients of oxidative damage, implies a
intracellular signaling (Tan and Finkel, 2020). As a Ca2+ buffer vicious cycle wherein impaired mitochondria produce increased
zone within cells, mitochondria collaborate with the endoplasmic amounts of ROS, in turn leading to a progressive accumulation of
reticulum, extracellular matrix, and other cellular structures by damage that culminates in the process of aging (Figure 1).
Controlling the Ca2+ concentration to modulate cellular activities In all cells, an intracellular balance between the production and
(Aoyama-Ishiwatari and Hirabayashi, 2021). Additionally, elimination of ROS, known as “redox homeostasis”, necessitates
mitochondria participate in the regulation of various cellular efficient coordination of reactions across various cells and is
stress responses, such as OS, heat stress, and apoptosis (Chien regulated by an intricate network of pro-oxidation and
et al., 2014; Yao et al., 2023). The proper execution of antioxidant systems (Chianese and Pierantoni, 2021). The latter,
mitochondrial functions counts on the structural the source of total antioxidant capacity, encompasses non-
compartmentalization, which allows mitochondria to respond to enzymatic scavengers, as well as detoxifying enzymes that operate
intracellular signals timely. Over the past decade, substantial within distinct organelles. Non-enzymatic antioxidants include
evidence has emerged, shedding light on the involvement of albumin, urate, taurine, hypotaurine, pyruvate, lactate,
mitochondria in gamete formation and embryonic development, tocopherol, ergothioneine, ascorbic acid and melatonin (Ball,
as well as the elevated prevalence of reproductive disorders, such as 2008; Gonzalez-Arto et al., 2016). The antioxidant enzyme
inflammation of genitourinary system (Jankauskas et al., 2018), defense system involves superoxide dismutase (SOD), catalase
asthenospermia (Li et al., 2023), and polycystic ovary syndrome (CAT), glutathione peroxidase (GPX), glutathione reductase
(Jiang et al., 2022). (GSR), peroxiredoxin (PRDX), and so on (Ben Abdallah et al.,
2009; Klopotowska et al., 2022). OS is defined as the imbalance
between the detoxification of cell antioxidant defense system and the
2.2 Mitochondria as the major source of ROS production of ROS. In the status of OS, extensive ROS is produced
generation and accumulate within the whole cell. ROS is extremely reactive and
can engage in reactions with virtually any substantial molecules,
In eukaryotes, the production of ATP occurs within leading to lipid peroxidation and DNA impairment, among other
mitochondria through oxidative metabolism of nutrients, which consequences (Yang et al., 2010). Polyunsaturated fatty acid
involves two key steps: OxPhos process (Iovine et al., 2021), and the moieties of phospholipids and cholesterol are preferred substrates
oxidation of nicotinamide adenine dinucleotide hydride (NADH) or for lipid hydroperoxides (e.g., Malonaldehyde and 4-
flavin adenine nucleotide phosphate (FADH2), or the β-oxidation of hydroxynonenal) and OH generated in membranes (Brouwers
fatty acids (Shen et al., 2017). The electron transport chain facilitates et al., 2005). Active ROS attacks directly sensitive DNA and
the oxidation of NADH and FADH2 generated through glycolysis damages components of the chromosome, such as telomeres,
and the TCA cycle. Simultaneously, it actively transports protons causing DNA strand breaks and altered repair mechanisms that
across the IMM, leading to the accumulation of protons in the IMS. can lead to the acquisition of genomic changes (Sallmyr et al., 2008).
This process establishes a transmembrane proton gradient with These impaired molecules disrupt regular cellular metabolism,
proton-motive force. Complex V (or called ATP synthase), prompting a subsequent escalation in ROS production, thus
harnesses the proton flow driven by this gradient to catalyze the establishing a detrimental cycle. Ultimately, the excessive
synthesis of ATP from ADP and phosphoric acid. Consequently, the accumulation of ROS culminates in oxidative damage to

Frontiers in Cell and Developmental Biology 03 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

macromolecules, cells, and even the overall structure and surveillance, repair, and proteolytic pathways, it is noteworthy
functionality of the organism (Redza-Dutordoir and Averill- that these defense systems are susceptible to age-related
Bates, 2016). impairments, thereby diminishing their capacity to effectively
eliminate proteins damaged by oxidation (Peters et al., 2020).
ROS-induced damage can impact the primary molecular
3 Effects of excessive ROS levels on constituents of oocytes, encompassing lipid membranes, nucleic
reproductive aging acids, and protein complexes, thereby disrupting protein
functionality and cellular equilibrium (Husain and Mahmood,
3.1 The role of ROS in oocyte quality and 2019; Cui et al., 2022). Increased ROS can also cause structural
female fertility deterioration of organelles, leading to mitochondrial dysfunction,
telomere shortening due to decreased telomerase activity,
ROS plays a dual role in oocyte development, serving as both a abnormalities in spindle assembly, and abnormal cytoskeletal
beneficial and detrimental factor. In its normal functioning, ROS dynamics (Zhang et al., 2018; Zhou et al., 2019). These abnormal
assumes a crucial role in regulating folliculogenesis, meiosis, conditions significantly diminish the effectiveness of meiotic
ovulation, and embryonic development by acting as secondary recombination, resulting in errors in chromosome segregation
messengers in cellular signaling pathways (Agarwal et al., 2012). and loss (Liu et al., 2004; Schatten and Sun, 2015; Yang et al.,
However, when present in excessive amounts, ROS will result in OS 2020). Consequently, oocytes undergo abnormal meiotic division
and exert detrimental effects including a series of reproductive and experience a substantial decline in egg quality (Figure 2).
diseases such as endometriosis and polycystic ovary syndrome The absence of antioxidant genes in the ovaries can disrupt
(Wang F. et al., 2023; Weng et al., 2023; Zhou et al., 2023). The follicular growth. Histological analysis conducted on the ovaries
initial proposition by Tarin (Tarin, 1995) regarding ovarian from SOD1 knockout mice demonstrated many primary and small
senescence posits that oocytes exhibit reduced resistance to ROS, antral follicles but few corpora lutea (Matzuk et al., 1998). Deletion
which is a significant contributor to oocyte impairment. The of SOD1 resulted in a defect in implantation of embryos to the wall
prolonged duration of meiosis-arrested oocytes increases their of the uterine horns and small litters, suggesting that SOD1 may play
vulnerability to oxidative damage, as indicated by the disturbance a critical role in germ cell survival (Ho et al., 1998). The gilts supplied
of protein homeostasis, particularly in terms of proteolysis (Wu for dietary 2-hydroxy-4-methylselenobutanoic acid showed
et al., 2022). Even though oocytes possess the capability to increased weight of uteri accompanied by increased the activity
counteract this effect through the activation of intricate of T-AOC, T-SOD, GPX, TXNRD and decreased MDA content in

FIGURE 2
Detrimental effects of excessive ROS and mitochondrial dysfunction on different reproduction stages from gamete formation to embryonic
development in the uterus. For the male, excessive ROS and mitochondrial dysfunction would cause inactive sperms with loss of fertility and declined
motility by decreased energy supplement to fail to sperm penetration. For the female, excessive ROS and mitochondrial dysfunction would influence the
entire oogenesis, increase the possibility of lessened quantity of ovum, and decline fertilization potential. Otherwise, excessive ROS can disturb the
fetal development and even occurs developmental disorders. A solid arrow indicates that the process is normal, and a dashed arrow indicates that the
process is defective.

Frontiers in Cell and Developmental Biology 04 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

granulosa cells (GCs), implied that the drug is conducive to and elevated levels of ROS and malondialdehyde, which can be rescued
promoting follicle development by antioxidant pathway (Lin by the antioxidants N-acetyl-L-cysteine or tocopherol (Chen et al.,
et al., 2022). Furthermore, many advantageous factors from 2022). These studies indicate that antioxidant enzymes in the sperm
environment like resveratrol can elevate the level of ROS to storage organ play a strong protective role against OS ensuring
generate various intracellular toxic effects including embryonic maintenance of sperm vitality and overall quality. Understanding the
stage cellular toxicity and teratogenicity in various mammalian regulatory mechanisms of the endogenous oxidative defense system,
species and even the impact on human fetal development which serves as the primary barrier against high OS in sperm, is of
(Manzo et al., 2010; Auti et al., 2022). In recent years, more and significant value for improving reproductive capacity.
more drugs have been evidenced playing a significantly antioxidant
role to help delay or rescue the aging phenotypes of reproduction
(Yang et al., 2022; Wang et al., 2023b; Wang et al., 2023c). For 4 Mitochondrial dysfunctions
instance, the antioxidant Salidroside can increase the number of associated with reproductive aging
blastocyst cells, the proliferation of blastocysts, and the expressions
of pluripotency genes while reducing ROS levels in oocytes and 4.1 Consequences of inadequate
enhancing intracellular GSH levels (Shi et al., 2023). Currently, mtDNA levels
screening of drugs to effectively decrease the level of ROS possesses a
broad prospect to ameliorate the quality of oocytes and their Mammalian mitochondria are naturally inherited through the
subsequent embryonic development. maternal lineage (Hutchison et al., 1974), whereas the sperm
mitochondria become degraded after fertilization (Sutovsky et al.,
1999). Hence, the determination of oocyte health, fertilization, and
3.2 The role of ROS in sperm functions and early embryonic development hinges on the abundance and quality of
testis health the mitochondrial population within the oocyte to a great extent (Wai
et al., 2010; Fragouli and Wells, 2015). The quality as well as the number
The decline in male reproductive function with age is a prevalent of mitochondria within the oocyte play significant roles as markers for
phenomenon observed in mammals, birds, and flies (Fricke and Koppik, oocyte quality and pivotal indicators for fertilization and embryo
2019). Research conducted on males have revealed that aging poses development (Tilly and Sinclair, 2013). During the process of oocyte
smaller testes (Mahmoud et al., 2003), a notable decrease in the average growth subsequent to follicle recruitment, the primordial oocyte
quantity of sertoli cells (Johnson et al., 1984), and lower testosterone experiences a substantial increase in the number of mitochondria
levels (Pines, 2011). A retrospective analysis of semen parameters (e.g., (Wassarman et al., 1976). Hence, the replication of mtDNA and
ejaculate volume, sperm concentration, and sperm motility), conducted mitochondrial biogenesis is sustained in parallel with the follicle
on a sample size exceeding 5,000 men aged 16–72 years indicates semen growth (Larsson et al., 1998; Hou et al., 2019). According to
parameters commence a decline after 34 years of age (Stone et al., 2013). estimations, there is a decline of approximately 4 copies of human
Sperm quality, as one of the decisive factors affecting animal mtDNA every 10 years, which has been associated with compromised
reproductive capacity, is crucial for the propagation across species. post-implantation development and an elevated likelihood of
The abnormal increase of ROS in the reproductive system by age is subfertility (Zhang et al., 2017; Busnelli et al., 2018). The analysis of
one of the core causes of the decline of sperm quality. an in vitro fertilization/intracytoplasmic sperm injection program
Once ROS levels increase abnormally, it can lead to decreased suggested the circulating level of anti-Müllerian hormone, a
fluidity of the mitochondrial membrane structure, protein denaturation, hormone essential to the regulation of ovarian follicle growth by
and increased membrane fragility within the sperm (Fleury et al., 2002). inhibiting initial follicle recruitment, is strongly correlated with
This disruption can directly result in the inability to maintain the proton mtDNA abundance within GCs (Bowolaksono et al., 2022). The
gradient across the inner mitochondrial membrane, causing the accumulating evidence from current studies exhibits that an
biochemical basis of ATP synthesis to be compromised. As a result, association exists between maternal age and the incidence of
sperm loses its motility and ability to move effectively due to decreased mtDNA deletion in oocytes and GCs (Keefe et al., 1995; Chan et al.,
energy levels (Chianese and Pierantoni, 2021). In order to cope with the 2005). Additionally, a greater number of mtDNA copies in the embryo
risk of high OS, sperm have developed a unique oxidative defense is linked to higher fecundity (Murakoshi et al., 2013; Babayev et al.,
mechanism to maintain low levels of ROS within their internal 2016). Generally, the inadequate amount of mtDNA is insufficient in
environment (Guerriero et al., 2014; O’flaherty, 2019). Besides supplying the required energy reserves for follicular growth,
providing a liquid environment for sperms, seminal plasma serves as consequently resulting in oocyte degeneration.
a source of numerous nutrients and protective substances that modulate
sperm capacitation and gamete interaction (Vashisht and Gahlay,
2023). Among multitudinous substances, antioxidant system is high- 4.2 Consequences of abnormal
profile for their roles involved in the process (Yin et al., 2023). In the mitochondrial dynamics for the
seminal plasma of jackasses, the activities of antioxidant enzymes (SOD, development of reproductive system
CAT, GPX, and GSR) were significantly correlated with sperm motility
and forward movement ability (Maehly and Chance, 1954; Wdowiak Mitochondria are organelles characterized by their dynamic
et al., 2015; Papas et al., 2019). The impairment by NiCl2 treatment on nature with an intricate network of tubular structures, displaying
sperm total motility and progressive motility in a dose- and time- an amazing plasticity of morphology and distribution.
dependent manner is characterized by interference with Ca2+ signaling Mitochondrial dynamics includes the fusion and fission between

Frontiers in Cell and Developmental Biology 05 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

the IBM and the OM during mitochondrial movement with embryonic genome may necessitate mitochondrial fusion to
surrounding mitochondria for exchange of matrix and membrane facilitate this significant developmental process. Inhibition of
components (Wai and Langer, 2016). The two pivotal events Drp1 in embryonic stem cells induced totipotency-to-
maintain a continuously coordinated cycle essential for pluripotency transition characterized by decreased activity of the
facilitating adequate local energy generation and distribution glycolysis/gluconeogenesis pathway by failed silencing of 2-cell
required by each cellular organelle to execute their respective genes in embryos, ultimately leading to impeded development of
functions (Frederick and Shaw, 2007). This enables mitochondria early embryos (Guo et al., 2023). High expression of Mfn1
to preserve its optimal functioning status and structural integrity significantly promoted early development of bovine embryos with
(Suen et al., 2008). Fusion serves to alleviate stress by amalgamating somatic cell nuclear transfer by increasing ATP level and
the constituents of partially impaired mitochondria, thereby mitochondrial membrane potential, while reducing H2O2
enhancing their functionality, while fission is necessary for the generation by improving OxPhos (Babayev et al., 2016).
elimination of compromised and less efficient mitochondria or Mitochondrial fission and fusion homeostasis are critical factors
for the generation of novel ones (Karbowski et al., 2002). ensuring the optimal mitochondrial activity in oocytes and embryos,
Assessing the impact of mitochondrial fusion and fission on thereby facilitating the normal development of embryos, and
cellular processes proves challenging due to the multifaceted preventing oxidative damage. With the identification and
nature of these processes, which can yield diverse outcomes validation of additional molecules or drugs, the manipulation of
dependent on the temporal and spatial factors of their mitochondrial dynamics may emerge as a prospective avenue for
occurrence. It is worth noting that mitochondria are highly improving reproductive health.
prevalent organelles in oocytes and embryos. Throughout
maturation of oocytes and embryonic development preceding
implantation, mitochondria undergo persistent and dynamic 4.3 Disruption of mitochondrial
modifications to facilitate essential cellular developmental events bioenergetics for oocyte maturation
(Sathananthan and Trounson, 2000; Harvey, 2019). The actional
nature of energy demands leads to corresponding fluctuations in It is well known that mitochondria are key regulators of
mitochondrial dynamics, which effectively and unitedly regulate structure formation, metabolic activity, and function (Sherratt,
metabolism to meet the material and energy requirements necessary 1991). Their most primary role, energy production, is critical for
for oocyte maturation and embryonic development (Schatten et al., function of organs with high metabolism like ovary and testis. The
2005; Zhao et al., 2016). oocyte possesses a greater quantity of mitochondria compared to
The involvement of mitochondrial dynamic-related proteins in other cells in mammals (Kim et al., 2019). Research using
germ cells and embryos has been investigated extensively in mammalian models has provided compelling evidence that
mammals. The novel evidence from mitofuscin 2 (Mfn2) mitochondria are imperative for the development of proficient
knockout mice indicates that the seminiferous tubules display a female gametes. During the maturation of follicles, as the
vacuous morphology due to impaired spermatogenesis and efficiency of glycolysis in the oocyte is limited due to low
increased apoptosis, parallel to an interference of cellular expression of phosphofructokinase (Leese and Barton, 1984),
respiration effected by lower expression of the subunits of there is a metabolic shift from glycolysis to OxPhos, which
OxPhos complexes (Wang X. et al., 2021). facilitates the production of increased ATP levels conducive to
Indeed, it has been empirically shown that, alongside supporting various processes such as cytoplasmic and nuclear
mitochondrial fusion, the process of fission is also necessary in maturation, spindle formation, and polar body extrusion
governing the preservation of early spermatogenesis (Honda and (Barbehenn et al., 1974; Zhang et al., 2006; Dumollard et al.,
Hirose, 2003). Stage-specific enhancement in dynamin related 2007b). In this stage, there is a significant increase in the
protein 1 (Drp1) expression in rat spermatids cooperated with abundance of functional mitochondria, which serves as the
Mfn2 to trigger the mitochondrial ubiquitination to attain their primary provider of ATP for the reorganization of cytoplasmic
homogenization by fusion and fission. The Drp1-deleted oocytes and nuclear activities, such as gene expression and the initiation of
displayed clustering of aberrant mitochondria, compromised crucial signaling cascades (Kirillova et al., 2021). Additionally, the
calcium homeostasis, impaired secretory function, multiorganelle spatial arrangement of mitochondria undergoes a transformation,
deformation, disrupted meiosis, and ultimately, age-dependent transitioning from a homogenous distribution throughout the
infertility in female mice (Udagawa et al., 2014). Mitofuscin 1 cytoplasm during the germinal vesicle stage to a clustered
(Mfn1) knockout oocytes exhibited impaired oocyte-GC aggregation near the perinuclear region in metaphase I or II
communication, downregulated cadherins and connexins, oocytes (Wang L. Y. et al., 2009).
resulting in follicle developmental arrest at the secondary follicle Previously, it has been documented that there is a notable rise in
stage as well as depletion of ovarian follicular reserve. These ATP level alongside the clustering of mitochondria in the
phenotypes could be partially rescued by treatment with perinuclear region during the maturation of oocytes in mice and
ceramide synthesis inhibitor myriocin (Zhang et al., 2019). pigs (Yu et al., 2010). Inefficient mitochondrial bioenergetics can
Targeted deletion of Mfn2 in mice showed reduced rates of contribute to a detrimental impact on the effectiveness of
oocyte maturation and fertilization, along with changes in bidirectional signal communications between GCs and oocytes,
mitochondrial allocation and spindle assembly (Liu et al., 2016), primarily mediated by the secreted sex hormones and
which highlighted the involvement of mitochondrial dynamics in intercellular gap junctions (Best et al., 2015). Consequently, this
regulating chromosome separation in oocytes. The activation of the disruption may result in impairments in oocyte maturation. In

Frontiers in Cell and Developmental Biology 06 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

addition, it has been demonstrated that a decline in ATP content the sex difference in lifespan expectancy: the accumulation of
generated through OxPhos process can impede the cytoplasmic mtDNA mutations may potentially make an adverse consequence
microtubule network, leading to a delayed or hindered maturation for males if they are neutral or advantageous for females (Innocenti
of oocytes in mice (Marques-da-Silva et al., 2011). Disruption of et al., 2011). These effects may manifest directly, such as through
OxPhos, the core process for ATP production, can cause halting of mitochondrial function (Anderson et al., 2022), or indirectly, such as
oocyte maturation, chromosomal misdistribution, and hindered retroegulation of nuclear genome by mtDNA (Hamalainen et al.,
embryo development (Takeuchi et al., 2005; Thouas et al., 2006; 2019). However, the substantial variations in the reproductive cycle
Wyman et al., 2008).The conditional disruption of the decaprenyl and the pace of reproductive senescence among males and females
diphosphate synthase subunit 2 (Pdss2) gene, leading to a deficiency have yet to be adequately elucidated and warrant further
of CoQ in oocytes, overviews numerous phenotypic alterations investigation.
typical of mitochondrial dysfunction in oocytes associated with
reproductive aging, and could be reversed by the administration
of CoQ10 (Ben-Meir et al., 2015). During the matured phase of 5.2 ROS and mitochondria interact in
oocyte, mitochondria undergo relocation to varying regions to fulfill ovarian aging
localized energy requirements. The occurrence of rapid increases in
ATP demand and subsequent mitochondrial redistribution are The female reproductive longevity is predominantly influenced
closely linked to the development of the oocyte (Figure 2). An by the ovarian health. A shortened reproductive longevity in female
adequate number of functional mitochondria makes up a decisive with diminished fertility or potential infertility can be attributed to
factor in determining the quality of the oocyte. the accelerated ovarian aging. Ovarian aging is characterized by a
decline in oocyte quality, as well as a decrease in the quantity of
follicles and oocytes. The theory of ovarian aging, initially proposed
5 Discussion by Tarin, posits that oocytes exhibit a reduced capacity to withstand
ROS, a crucial factor contributing to oocyte impairment (Tarin,
5.1 Variations in the rates of reproductive 1995). At a fundamental level, ROS functions as a secondary
aging exist between males and females messenger in cellular signaling and assumes a crucial role in
facilitating folliculogenesis, meiosis, and ovulation processes (Ellis
In mammals, the process of reproduction can be categorized into et al., 1987). However, the detrimental effects of excessive ROS can
distinct stages, namely, sex differentiation, sexual maturation, germ adversely affect the fundamental molecular components of oocytes,
cell development encompassing spermatogenesis and oogenesis, leading to the disruption of protein functionality and cellular
mating, fertilization, pregnancy, and parturition. The lifespan homeostasis (e.g., telomerase) (Sasaki et al., 2019). The two hit
expenses of sustaining reproductive capability, and of “telomere theory of reproductive aging” proposed that oocytes
reproduction itself primarily focus on maintaining a stable experience a greater accumulation of environmental and
developmental environment and ensuring an adequate energy endogenous oxidative damage throughout the lifetime of female
supply, as any deviation or mistake at any stage can impede individuals, which could be mediated by telomeres (Robinson et al.,
successful reproduction (Zhang and Hood, 2016). The 2023). Telomeres undergo elongation through two distinct
completion of reproduction necessitates the collaborative efforts mechanisms, telomerase and alternative lengthening of telomeres.
of both females and males, although females typically assume greater Alternative lengthening of telomeres is infrequently observed except
responsibilities, such as embryonic development, parturition, and telomeropathy. Sperms harbor the longest telomeres in the body,
lactation. Intriguingly, in the context of human reproduction, men which are continuously generated by a telomerase-active progenitor,
exhibit a prolonged fertility span throughout their lives, consistently spermatogonia, throughout the male’s lifespan. In contrast, the
generating sperm, whereas women experience a comparatively telomerase activity in the female germline is initially low and
limited window of fertility. The decline in female fertility decreases following the initiation of meiotic arrest (Liu et al.,
commences around the age range of 30–35, manifesting in an 2007; Jeon et al., 2022). Otherwise, telomere attrition may still
augmented likelihood of miscarriage, diminished chances of occur due to the presence of ROS (Kordowitzki, 2021).
conception, and an expedited progression of reproductive Therefore, oocytes frequently carry short telomeres trapped into
senescence, characterized by an increased probability of oocyte a risky survival situation. Even though this theory has some
aneuploidy (Mogessie, 2020). The different reproductive aging rationality, ROS may interact with the health of oocytes in more
patterns between sexes may be associated with the global aspects than just telomere length. The disruption caused by ROS will
phenomenon that women outlive men worldwide (Thornton, trigger endoplasmic reticulum stress, autophagy, and proteasome
2019). Sex differences in life expectancy also share a a prevalent dysfunction, culminating in follicular atresia and a decline in both
trend in many species, including mammals (Hoffman et al., 2013), the quality and quantity of oocytes (Sasaki et al., 2019). Furthermore,
birds (Liker and Szekely, 2005), and insects (Bateman, 1948). Males mitochondrial quality determines the potential of oocyte fertility
may be sacrificing access to resources to longer lifespan expectancy and the developing embryo (Dumollard et al., 2007a). Mitochondrial
in favor of sustaining an extended reproductive potential. One dysfunction can give rise to heightened ROS level and the
explanation for trade-offs between longevity and reproduction is accumulation of damaged age-related proteins (Seli et al., 2019).
that male sex hormones reduce the lifespan of men, proved by a Excessive ROS levels can prompt intracellular OS and contribute to a
longer lifespan in castrated men (Min et al., 2012). Asymmetric decline in the quantity of primordial follicles and the mortality of
inheritance of mtDNA presents a novel perspective for explaining pre-implantation embryos, consequently diminishing the

Frontiers in Cell and Developmental Biology 07 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

reproductive lifespan of females (Sasaki et al., 2019). The interplay transgenic mice showed that sperm mitochondria are not
between ROS, mtDNA damage, and mitochondrial dysfunction subjected to elimination by the early embryonic degradation
establishes a vicious cycle (Figure 1), the occurrence of which can mechanisms, ubiquitin-proteasome, and autophagy. Instead,
initiate a cascading effect-an increase in ROS levels and mtDNA sperm mitochondria may persist uniformly in certain cells
mutagenesis, ultimately resulting in a decline in until the morula stage, suggesting paternal mtDNA could be
reproductive capacity. transmitted to offspring (Luo et al., 2013). Indisputably,
mitochondria play a crucial role in the processes of sperm
fertility and fertilization, and any aberration within the
5.3 The role of paternal mitochondria in mitochondria has the potential to cause impairment of sperm
offspring development fertility and subsequent male infertility.

Although reproductive aging in males may not receive as


much attention as in females, it similarly leads to infertility and 5.4 ROS and mitochondria are therapeutic
has detrimental effects on offspring. The gradual decline in male targets for improvement of
reproductive function with age is a widespread phenomenon reproductive aging
observed across species (Fricke and Koppik, 2019). Research in
males has identified smaller testes, a notable reduction in the The association between mitochondrial function and ROS is
average number of Sertoli cells, and decreased testosterone levels particularly pronounced in the context of reproductive aging.
as consequences of aging (Mahmoud et al., 2003). During the Currently, strategies aimed at mitigating oxidative damage and
differentiation of mammalian sperms, a significant portion of the enhancing mitochondrial quality control have emerged as
cytoplasm is eliminated; however, a subset of mitochondria prominent therapeutic approaches for addressing reproductive
persists within the sperms, assuming a tubular arrangement in aging and related disorders in clinical settings. The measurement
proximity to the middle flagellum (Amaral et al., 2013). of ROS levels in follicular fluid can serve as a biochemical marker for
Furthermore, OS has been associated with male reproductive assessing follicular metabolic age (Wang et al., 2012), and male
disorders (Lou et al., 2006; Mansouri Torghabeh et al., 2022; Xu infertility (Wang et al., 2003). Some natural compounds with
et al., 2022). A harmonious redox state as well as normal antioxidant properties that are isolated from medicinal plants
mitochondria are crucial to support spermatogenesis, such as resveratrol, have also been shown to protect against
epididymal transport, and subsequent activities following ovarian aging through multiple mechanisms, including
ejaculation (motility, capacitation, and acrosome reaction) counteracting cytotoxicity (Banu et al., 2016), preventing the loss
(Pelliccione et al., 2011). When the balance is disrupted, the of the GCs (Ozatik et al., 2020), improving renewal capacity of
resulting OS will adversely affect sperms in various aspects such oogonial stem cells (Wu et al., 2019), and diminishing ovarian
as the genome, epigenome, lipidome, and proteome, thereby inflammation (Said et al., 2016). Mitochondrial membrane
contributing to male infertility (Wang et al., 2003; Lim et al., potential and mtDNA copy number have been identified as
2019). In most instances, the inheritance of mitochondria occurs potential biomarkers for the selection of superior embryos and
maternally, whereas paternal mitochondria are eliminated. improved pregnancy (Wang J. et al., 2021) as well as for
Nevertheless, the precise mechanisms responsible for the enhancing the accuracy of diagnosing male infertility (Karimian
selective paternal mitochondrial elimination, remain largely and Babaei, 2020). Experts in reproductive medicine have proposed
elusive. It is traditionally considered that sperm mitochondria that mitochondrial transplantation could serve as an innovative
and their mtDNA should be removed early in the embryo by approach to rejuvenating oocyte quality and addressing age-related
selective mitophagy to protect the embryo and offspring from infertility or recurrent in vitro fertilization failures (Kristensen et al.,
paternal mtDNA damage due to vulnerability of sperm mtDNA 2017; Labarta et al., 2019). This technique has demonstrated positive
to oxidative damage (Marchetti et al., 2002). Paternal outcomes in humans and various animal species. The transfer of
mitochondria elimination is carried out through the autologous or heterologous mitochondria successfully enhanced
involvement of autophagy receptors in ubiquitin-dependent oocyte quality and improved in vitro fertilization results in a
and ubiquitin-independent manners (Wei et al., 2017). For bovine embryo model (Srirattana and St John, 2018). In aged
example, an allophagy receptor, ALLO-1, accumulates on the mice, MII oocytes were fertilized with significantly increased
paternal organelles and leads to leads to local autophagosome blastocyst ratio after intracytoplasmic sperm injection combined
formation depending on the ubiquitin modification after with mitochondria from endometrial mesenchymal stem cells
fertilization (Sato et al., 2018). FNDC-1, the Caenorhabditis (Zhang et al., 2023). Nevertheless, it is important to acknowledge
elegans ortholog to human FUNDC1 (FUN14 domain that this technique is limited in its ability to effectively enhance
containing1), is a ubiquitin-independent mitophagy receptor blastocyst quality (Labarta et al., 2019). Furthermore, the safety and
on the OMM involved in mitochondrial quality control under ethical considerations surrounding the manipulation of
hypoxic conditions, and plays a crucial role in selective mitochondria and blastocyst quality necessitate a more
degradation of paternal mitochondria by the asymmetric high- comprehensive understanding of their interaction. In essence,
expression on sperm mitochondria (Lim et al., 2019). understanding the molecular processes that govern the balance of
Nevertheless, the role of sperm mitochondria in the ROS and mitochondrial function is crucial for maintaining
fertilization and their final fate after fertilization remain reproductive health. This knowledge is essential for identifying
subjects of ongoing debate. Recent investigations conducted in key targets to improve the quality of gametes. By doing so, we

Frontiers in Cell and Developmental Biology 08 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

can delve into the causes of reproductive aging and disorders, paving Funding
the way for effective therapeutic strategies.
The author(s) declare financial support was received for the
research, authorship, and/or publication of this article. This work
6 Future perspectives was supported by the National Natural Science Foundation of China
(Nos. 32272939 and 31830094, and 31902215); Natural Science
The factors driving diverse aging phenotypes, especially Foundation of Chongqing, China (No. cstc2021jcyj-cxtt0005);
concerning reproductive aging, are expected to be complex, High-level Talents Program of Southwest University
involving changes across various physiological systems. Although (SWURC2021001); and China Agriculture Research System of
we can differentiate between extrinsic and intrinsic factors, it is likely MOF and MARA (No. CARS-18-ZJ0102).
that the levels of ROS and mitochondrial quality associated with
reproductive aging are influenced by both forces, suggesting a
significant interplay between them. Delving into the intricate
interplay among ROS, mitochondria, and germline function will
Conflict of interest
yield valuable insights into underlying molecular mechanism of
The authors declare that the research was conducted in the
reproductive aging. Further exploration is also conducive to lay the
absence of any commercial or financial relationships that could be
groundwork to address fertility issues in humans by exploring
construed as a potential conflict of interest.
potential strategies from the point of ROS level and mitochondria.

Author contributions Publisher’s note


JS: Conceptualization, Writing–original draft, Writing–review All claims expressed in this article are solely those of the authors
and editing. LX: Writing–original draft, Writing–review and editing. and do not necessarily represent those of their affiliated
ZZ: Visualization, Writing–review and editing. YW: Writing–review organizations, or those of the publisher, the editors and the
and editing. PK: Writing–review and editing. LR: Writing–review reviewers. Any product that may be evaluated in this article, or
and editing. FD: Conceptualization, Funding acquisition, claim that may be made by its manufacturer, is not guaranteed or
Supervision, Validation, Writing–review and editing. endorsed by the publisher.

References
Agarwal, A., Aponte-Mellado, A., Premkumar, B. J., Shaman, A., and Gupta, S. (2012). Ben Abdallah, F., Dammak, I., Mallek, Z., Attia, H., Hentati, B., and Ammar-Keskes,
The effects of oxidative stress on female reproduction: a review. Reprod. Biol. L. (2009). Effects of date seed oil on testicular antioxidant enzymes and epididymal
Endocrinol. 10, 49. doi:10.1186/1477-7827-10-49 sperm characteristics in male mice. Andrologia 41, 229–234. doi:10.1111/j.1439-0272.
2009.00923.x
Amaral, A., Lourenco, B., Marques, M., and Ramalho-Santos, J. (2013). Mitochondria
functionality and sperm quality. Reproduction 146, R163–R174. doi:10.1530/REP-13-0178 Ben-Meir, A., Burstein, E., Borrego-Alvarez, A., Chong, J., Wong, E., Yavorska, T.,
et al. (2015). Coenzyme Q10 restores oocyte mitochondrial function and fertility during
Anderson, L., Camus, M. F., Monteith, K. M., Salminen, T. S., and Vale, P. F. (2022).
reproductive aging. Aging Cell. 14, 887–895. doi:10.1111/acel.12368
Variation in mitochondrial DNA affects locomotor activity and sleep in Drosophila
melanogaster. Hered. (Edinb) 129, 225–232. doi:10.1038/s41437-022-00554-w Best, M. W., Wu, J., Pauli, S. A., Kane, M. A., Pierzchalski, K., Session, D. R., et al.
(2015). A role for retinoids in human oocyte fertilization: regulation of connexin 43 by
Aoyama-Ishiwatari, S., and Hirabayashi, Y. (2021). Endoplasmic reticulum-
retinoic acid in cumulus granulosa cells. Mol. Hum. Reprod. 21, 527–534. doi:10.1093/
mitochondria contact sites-emerging intracellular signaling hubs. Front. Cell. Dev.
molehr/gav017
Biol. 9, 653828. doi:10.3389/fcell.2021.653828
Blanchet, L., Buydens, M. C., Smeitink, J. A., Willems, P. H., and Koopman, W. J.
Archie, E. A., Altmann, J., and Alberts, S. C. (2014). Costs of reproduction in a long-
(2011). Isolated mitochondrial complex I deficiency: explorative data analysis of patient
lived female primate: injury risk and wound healing. Behav. Ecol. Sociobiol. 68,
cell parameters. Curr. Pharm. Des. 17, 4023–4033. doi:10.2174/138161211798764870
1183–1193. doi:10.1007/s00265-014-1729-4
Bowolaksono, A., Sundari, A. M., Fauzi, M., Maidarti, M., Wiweko, B., Mutia, K., et al.
Auti, A., Alessio, N., Ballini, A., Dioguardi, M., Cantore, S., Scacco, S., et al. (2022).
(2022). Anti-Mullerian hormone independently affect mtDNA copy number in human
Protective effect of resveratrol against hypoxia-induced neural oxidative stress. J. Pers.
granulosa cells. J. Ovarian Res. 15, 111. doi:10.1186/s13048-022-01047-4
Med. 12, 1202. doi:10.3390/jpm12081202
Brouwers, J. F., Silva, P. F., and Gadella, B. M. (2005). New assays for detection and
Babayev, E., Wang, T., Szigeti-Buck, K., Lowther, K., Taylor, H. S., Horvath, T., et al.
localization of endogenous lipid peroxidation products in living boar sperm after BTS dilution
(2016). Reproductive aging is associated with changes in oocyte mitochondrial
or after freeze-thawing. Theriogenology 63, 458–469. doi:10.1016/j.theriogenology.2004.09.046
dynamics, function, and mtDNA quantity. Maturitas 93, 121–130. doi:10.1016/j.
maturitas.2016.06.015 Busnelli, A., Lattuada, D., Rossetti, R., Paffoni, A., Persani, L., Fedele, L., et al. (2018).
Mitochondrial DNA copy number in peripheral blood: a potential non-invasive
Ball, B. A. (2008). Oxidative stress, osmotic stress and apoptosis: impacts on sperm
biomarker for female subfertility. J. Assist. Reprod. Genet. 35, 1987–1994. doi:10.
function and preservation in the horse. Anim. Reprod. Sci. 107, 257–267. doi:10.1016/j.
1007/s10815-018-1291-5
anireprosci.2008.04.014
Chan, C. C., Liu, V. W., Lau, E. Y., Yeung, W. S., Ng, E. H., and Ho, P. C. (2005).
Banu, S. K., Stanley, J. A., Sivakumar, K. K., Arosh, J. A., and Burghardt, R. C. (2016).
Mitochondrial DNA content and 4977 bp deletion in unfertilized oocytes. Mol. Hum.
Resveratrol protects the ovary against chromium-toxicity by enhancing endogenous
Reprod. 11, 843–846. doi:10.1093/molehr/gah243
antioxidant enzymes and inhibiting metabolic clearance of estradiol. Toxicol. Appl.
Pharmacol. 303, 65–78. doi:10.1016/j.taap.2016.04.016 Chen, C., Li, B., Huang, R., Dong, S., Zhou, Y., Song, J., et al. (2022). Involvement of
Ca(2+) and ROS signals in nickel-impaired human sperm function. Ecotoxicol. Environ.
Barbehenn, E. K., Wales, R. G., and Lowry, O. H. (1974). The explanation for the
Saf. 231, 113181. doi:10.1016/j.ecoenv.2022.113181
blockade of glycolysis in early mouse embryos. Proc. Natl. Acad. Sci. U. S. A. 71,
1056–1060. doi:10.1073/pnas.71.4.1056 Chianese, R., and Pierantoni, R. (2021). Mitochondrial reactive oxygen species (ROS)
production alters sperm quality. Antioxidants (Basel) 10, 92. doi:10.3390/
Bateman, A. J. (1948). Intra-sexual selection in Drosophila. Hered. (Edinb) 2,
antiox10010092
349–368. doi:10.1038/hdy.1948.21

Frontiers in Cell and Developmental Biology 09 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

Chien, C. Y., Chien, C. T., and Wang, S. S. (2014). Progressive thermopreconditioning Hutchison, C. A., Newbold, J. E., Potter, S. S., and Edgell, M. H. (1974). Maternal
attenuates rat cardiac ischemia/reperfusion injury by mitochondria-mediated inheritance of mammalian mitochondrial DNA. Nature 251, 536–538. doi:10.1038/
antioxidant and antiapoptotic mechanisms. J. Thorac. Cardiovasc Surg. 148, 251536a0
705–713. doi:10.1016/j.jtcvs.2013.12.065
Innocenti, P., Morrow, E. H., and Dowling, D. K. (2011). Experimental evidence
Cui, X., Zhang, Y., Lu, Y., and Xiang, M. (2022). ROS and endoplasmic reticulum supports a sex-specific selective sieve in mitochondrial genome evolution. Science 332,
stress in pulmonary disease. Front. Pharmacol. 13, 879204. doi:10.3389/fphar.2022. 845–848. doi:10.1126/science.1201157
879204
Iovine, J. C., Claypool, S. M., and Alder, N. N. (2021). Mitochondrial
Drewry, M. D., Williams, J. M., and Hatle, J. D. (2011). Life-extending dietary compartmentalization: emerging themes in structure and function. Trends Biochem.
restriction and ovariectomy result in similar feeding rates but different physiologic Sci. 46, 902–917. doi:10.1016/j.tibs.2021.06.003
responses in grasshoppers. Exp. Gerontol. 46, 781–786. doi:10.1016/j.exger.2011.06.003
Jankauskas, S. S., Silachev, D. N., Andrianova, N. V., Pevzner, I. B., Zorova, L. D.,
Drori, D., and Folman, Y. (1976). Environmental effects on longevity in the male rat: Popkov, V. A., et al. (2018). Aged kidney: can we protect it? Autophagy, mitochondria
exercise, mating, castration and restricted feeding. Exp. Gerontol. 11, 25–32. doi:10. and mechanisms of ischemic preconditioning. Cell. Cycle 17, 1291–1309. doi:10.1080/
1016/0531-5565(76)90007-3 15384101.2018.1482149
Dumollard, R., Duchen, M., and Carroll, J. (2007a). The role of mitochondrial Jeon, H. J., Kang, M., Kim, J. S., and Oh, J. S. (2022). TCTP overexpression reverses
function in the oocyte and embryo. Curr. Top. Dev. Biol. 77, 21–49. doi:10.1016/ age-associated telomere attrition by upregulating telomerase activity in mouse oocytes.
S0070-2153(06)77002-8 J. Cell. Physiol. 237, 833–845. doi:10.1002/jcp.30557
Dumollard, R., Ward, Z., Carroll, J., and Duchen, M. R. (2007b). Regulation of redox Jiang, X. L., Tai, H., Xiao, X. S., Zhang, S. Y., Cui, S. C., Qi, S. B., et al. (2022).
metabolism in the mouse oocyte and embryo. Development 134, 455–465. doi:10.1242/ Cangfudaotan decoction inhibits mitochondria-dependent apoptosis of granulosa cells
dev.02744 in rats with polycystic ovarian syndrome. Front. Endocrinol. (Lausanne) 13, 962154.
doi:10.3389/fendo.2022.962154
Ellis, L., Morgan, D. O., Jong, S. M., Wang, L. H., Roth, R. A., and Rutter, W. J. (1987).
Heterologous transmembrane signaling by a human insulin receptor-v-ros hybrid in Johnson, L., Zane, R. S., Petty, C. S., and Neaves, W. B. (1984). Quantification of the
Chinese hamster ovary cells. Proc. Natl. Acad. Sci. U. S. A. 84, 5101–5105. doi:10.1073/ human Sertoli cell population: its distribution, relation to germ cell numbers, and age-
pnas.84.15.5101 related decline. Biol. Reprod. 31, 785–795. doi:10.1095/biolreprod31.4.785
Fleury, C., Mignotte, B., and Vayssiere, J. L. (2002). Mitochondrial reactive oxygen Karbowski, M., Lee, Y. J., Gaume, B., Jeong, S. Y., Frank, S., Nechushtan, A., et al.
species in cell death signaling. Biochimie 84, 131–141. doi:10.1016/s0300-9084(02) (2002). Spatial and temporal association of Bax with mitochondrial fission sites, Drp1,
01369-x and Mfn2 during apoptosis. J. Cell. Biol. 159, 931–938. doi:10.1083/jcb.200209124
Fragouli, E., and Wells, D. (2015). Mitochondrial DNA assessment to determine Karimian, M., and Babaei, F. (2020). Large-scale mtDNA deletions as genetic
oocyte and embryo viability. Semin. Reprod. Med. 33, 401–409. doi:10.1055/s-0035- biomarkers for susceptibility to male infertility: a systematic review and meta-
1567821 analysis. Int. J. Biol. Macromol. 158, 85–93. doi:10.1016/j.ijbiomac.2020.04.216
Frederick, R. L., and Shaw, J. M. (2007). Moving mitochondria: establishing Keefe, D. L., Niven-Fairchild, T., Powell, S., and Buradagunta, S. (1995).
distribution of an essential organelle. Traffic 8, 1668–1675. doi:10.1111/j.1600-0854. Mitochondrial deoxyribonucleic acid deletions in oocytes and reproductive aging in
2007.00644.x women. Fertil. Steril. 64, 577–583. doi:10.1016/s0015-0282(16)57796-6
Fricke, C., and Koppik, M. (2019). Male reproductive ageing: a tale of the whole Khan, S. A., Reed, L., Schoolcraft, W. B., Yuan, Y., and Krisher, R. L. (2023). Control of
ejaculate. Reproduction 158, R219–R229. doi:10.1530/REP-18-0579 mitochondrial integrity influences oocyte quality during reproductive aging. Mol. Hum.
Reprod. 29, gaad028. doi:10.1093/molehr/gaad028
Gonzalez-Arto, M., Vicente-Carrillo, A., Martinez-Pastor, F., Fernandez-Alegre, E.,
Roca, J., Miro, J., et al. (2016). Melatonin receptors MT1 and MT2 are expressed in Kim, H. R., Won, S. J., Fabian, C., Kang, M. G., Szardenings, M., and Shin, M. G.
spermatozoa from several seasonal and nonseasonal breeder species. Theriogenology 86, (2015). Mitochondrial DNA aberrations and pathophysiological implications in
1958–1968. doi:10.1016/j.theriogenology.2016.06.016 hematopoietic diseases, chronic inflammatory diseases, and cancers. Ann. Lab. Med.
35, 1–14. doi:10.3343/alm.2015.35.1.1
Guerriero, G., Trocchia, S., Abdel-Gawad, F. K., and Ciarcia, G. (2014). Roles of
reactive oxygen species in the spermatogenesis regulation. Front. Endocrinol. Kim, K. B., Dunn, C. T., and Park, K. S. (2019). Recent progress in mapping the
(Lausanne) 5, 56. doi:10.3389/fendo.2014.00056 emerging landscape of the small-cell lung cancer genome. Exp. Mol. Med. 51, 1–13.
doi:10.1038/s12276-019-0349-5
Gumienny, T. L., Lambie, E., Hartwieg, E., Horvitz, H. R., and Hengartner, M. O.
(1999). Genetic control of programmed cell death in the Caenorhabditis elegans Kirillova, A., Smitz, J. E. J., Sukhikh, G. T., and Mazunin, I. (2021). The role of
hermaphrodite germline. Development 126, 1011–1022. doi:10.1242/dev.126.5.1011 mitochondria in oocyte maturation. Cells 10, 2484. doi:10.3390/cells10092484
Guo, S. M., Zhang, Y. R., Ma, B. X., Zhou, L. Q., and Yin, Y. (2023). Regulation of Klopotowska, M., Bajor, M., Graczyk-Jarzynka, A., Kraft, A., Pilch, Z., Zhylko, A.,
cleavage embryo genes upon DRP1 inhibition in mouse embryonic stem cells. Front. et al. (2022). PRDX-1 supports the survival and antitumor activity of primary and CAR-
Cell. Dev. Biol. 11, 1191797. doi:10.3389/fcell.2023.1191797 modified NK cells under oxidative stress. Cancer Immunol. Res. 10, 228–244. doi:10.
1158/2326-6066.CIR-20-1023
Hamalainen, R. H., Landoni, J. C., Ahlqvist, K. J., Goffart, S., Ryytty, S., Rahman, M.
O., et al. (2019). Defects in mtDNA replication challenge nuclear genome stability Koopman, W. J., Verkaart, S., Visch, H. J., Van Emst-De Vries, S., Nijtmans, L. G.,
through nucleotide depletion and provide a unifying mechanism for mouse progerias. Smeitink, J. A., et al. (2007). Human NADH:ubiquinone oxidoreductase deficiency:
Nat. Metab. 1, 958–965. doi:10.1038/s42255-019-0120-1 radical changes in mitochondrial morphology? Am. J. Physiol. Cell. Physiol. 293,
C22–C29. doi:10.1152/ajpcell.00194.2006
Harman, D. (1956). Aging: a theory based on free radical and radiation chemistry.
J. Gerontol. 11, 298–300. doi:10.1093/geronj/11.3.298 Kordowitzki, P. (2021). Oxidative stress induces telomere dysfunction and shortening
in human oocytes of advanced age donors. Cells, 10. doi:10.3390/cells10081866
Harvey, A. J. (2019). Mitochondria in early development: linking the microenvironment,
metabolism and the epigenome. Reproduction 157, R159–R179. doi:10.1530/REP-18-0431 Kristensen, S. G., Pors, S. E., and Andersen, C. Y. (2017). Improving oocyte quality by
transfer of autologous mitochondria from fully grown oocytes. Hum. Reprod. 32,
Heldt, H. W. (1972). Energy metabolism in mitochondria. Angew. Chem. Int. Ed.
725–732. doi:10.1093/humrep/dex043
Engl. 11, 792–798. doi:10.1002/anie.197207921
Labarta, E., De Los Santos, M. J., Escriba, M. J., Pellicer, A., and Herraiz, S. (2019).
Ho, Y. S., Gargano, M., Cao, J., Bronson, R. T., Heimler, I., and Hutz, R. J. (1998).
Mitochondria as a tool for oocyte rejuvenation. Fertil. Steril. 111, 219–226. doi:10.1016/
Reduced fertility in female mice lacking copper-zinc superoxide dismutase. J. Biol.
j.fertnstert.2018.10.036
Chem. 273, 7765–7769. doi:10.1074/jbc.273.13.7765
Larsson, N. G., Wang, J., Wilhelmsson, H., Oldfors, A., Rustin, P., Lewandoski, M.,
Hoffman, J. M., Creevy, K. E., and Promislow, D. E. (2013). Reproductive capability is
et al. (1998). Mitochondrial transcription factor A is necessary for mtDNA maintenance
associated with lifespan and cause of death in companion dogs. PLoS One 8, e61082.
and embryogenesis in mice. Nat. Genet. 18, 231–236. doi:10.1038/ng0398-231
doi:10.1371/journal.pone.0061082
Lee, H. C., and Wei, Y. H. (2001). Mitochondrial alterations, cellular response to
Holmstrom, K. M., and Finkel, T. (2014). Cellular mechanisms and physiological
oxidative stress and defective degradation of proteins in aging. Biogerontology 2,
consequences of redox-dependent signalling. Nat. Rev. Mol. Cell. Biol. 15, 411–421.
231–244. doi:10.1023/a:1013270512172
doi:10.1038/nrm3801
Leese, H. J., and Barton, A. M. (1984). Pyruvate and glucose uptake by mouse
Honda, S., and Hirose, S. (2003). Stage-specific enhanced expression of mitochondrial
ova and preimplantation embryos. J. Reprod. Fertil. 72, 9–13. doi:10.1530/jrf.0.
fusion and fission factors during spermatogenesis in rat testis. Biochem. Biophys. Res.
0720009
Commun. 311, 424–432. doi:10.1016/j.bbrc.2003.10.008
Li, J., Xu, X., Liu, J., Zhang, S., Wang, T., Liu, Y., et al. (2023). The immunity-related
Hou, X., Zhu, S., Zhang, H., Li, C., Qiu, D., Ge, J., et al. (2019). Mitofusin1 in oocyte is
GTPase IRGC mediates interaction between lipid droplets and mitochondria to
essential for female fertility. Redox Biol. 21, 101110. doi:10.1016/j.redox.2019.101110
facilitate sperm motility. FEBS Lett. 597, 1595–1605. doi:10.1002/1873-3468.14640
Husain, N., and Mahmood, R. (2019). Copper(II) generates ROS and RNS, impairs
Liker, A., and Szekely, T. (2005). Mortality costs of sexual selection and parental care
antioxidant system and damages membrane and DNA in human blood cells. Environ.
in natural populations of birds. Evolution 59, 890–897. doi:10.1554/04-560
Sci. Pollut. Res. Int. 26, 20654–20668. doi:10.1007/s11356-019-05345-1

Frontiers in Cell and Developmental Biology 10 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

Lim, H. Y., Ho, Q. S., Low, J., Choolani, M., and Wong, K. P. (2011). Respiratory are higher in jackasses than in stallions and are correlated with sperm motility in
competent mitochondria in human ovarian and peritoneal cancer. Mitochondrion 11, jackasses. Theriogenology 140, 180–187. doi:10.1016/j.theriogenology.2019.08.032
437–443. doi:10.1016/j.mito.2010.12.015
Pelliccione, F., Micillo, A., Cordeschi, G., D’angeli, A., Necozione, S., Gandini, L., et al.
Lim, Y., Rubio-Pena, K., Sobraske, P. J., Molina, P. A., Brookes, P. S., Galy, V., et al. (2011). Altered ultrastructure of mitochondrial membranes is strongly associated with
(2019). Fndc-1 contributes to paternal mitochondria elimination in C. elegans. Dev. Biol. unexplained asthenozoospermia. Fertil. Steril. 95, 641–646. doi:10.1016/j.fertnstert.
454, 15–20. doi:10.1016/j.ydbio.2019.06.016 2010.07.1086
Lin, H. Y., Yang, Y. N., Chen, Y. F., Huang, T. Y., Crawford, D. R., Chuang, H. Y., et al. Peters, A. E., Mihalas, B. P., Bromfield, E. G., Roman, S. D., Nixon, B., and Sutherland,
(2022). 2,3,5,4’-Tetrahydroxystilbene-2-O-β-D-Glucoside improves female ovarian J. M. (2020). Autophagy in female fertility: a role in oxidative stress and aging. Antioxid.
aging. Front. Cell. Dev. Biol. 10, 862045. doi:10.3389/fcell.2022.862045 Redox Signal 32, 550–568. doi:10.1089/ars.2019.7986
Liu, L., Bailey, S. M., Okuka, M., Munoz, P., Li, C., Zhou, L., et al. (2007). Telomere Pines, A. (2011). Male menopause: is it a real clinical syndrome? Climacteric 14,
lengthening early in development. Nat. Cell. Biol. 9, 1436–1441. doi:10.1038/ncb1664 15–17. doi:10.3109/13697137.2010.507442
Liu, L., Franco, S., Spyropoulos, B., Moens, P. B., Blasco, M. A., and Keefe, D. L. Redza-Dutordoir, M., and Averill-Bates, D. A. (2016). Activation of apoptosis
(2004). Irregular telomeres impair meiotic synapsis and recombination in mice. Proc. signalling pathways by reactive oxygen species. Biochim. Biophys. Acta 1863,
Natl. Acad. Sci. U. S. A. 101, 6496–6501. doi:10.1073/pnas.0400755101 2977–2992. doi:10.1016/j.bbamcr.2016.09.012
Liu, Q., Kang, L., Wang, L., Zhang, L., and Xiang, W. (2016). Mitofusin 2 regulates the Robinson, L. G., Kalmbach, K., Sumerfield, O., Nomani, W., Wang, F., Liu, L., et al.
oocytes development and quality by modulating meiosis and mitochondrial function. (2023). Telomere dynamics and reproduction. Fertil. Steril. 121, 4–11. doi:10.1016/j.
Sci. Rep. 6, 30561. doi:10.1038/srep30561 fertnstert.2023.11.012
Loose, J. A., Amrit, F. R. G., Patil, T., Yanowitz, J. L., and Ghazi, A. (2022). Meiotic Roger, A. J., Munoz-Gomez, S. A., and Kamikawa, R. (2017). The origin and
dysfunction accelerates somatic aging in Caenorhabditis elegans. Aging Cell. 21, e13716. diversification of mitochondria. Curr. Biol. 27, R1177–R1192. doi:10.1016/j.cub.2017.
doi:10.1111/acel.13716 09.015
Lou, J. G., Dong, J., Zheng, Y. C., Zhang, S. M., Xiao, W. Q., and Zhou, J. F. (2006). Said, R. S., El-Demerdash, E., Nada, A. S., and Kamal, M. M. (2016). Resveratrol
Increased oxidative stress and damage in patients with chronic bacterial prostatitis. inhibits inflammatory signaling implicated in ionizing radiation-induced premature
Biomed. Environ. Sci. 19, 481–486. doi:10.1111/j.1467-842X.2006.tb00794.x ovarian failure through antagonistic crosstalk between silencing information regulator 1
(SIRT1) and poly(ADP-ribose) polymerase 1 (PARP-1). Biochem. Pharmacol. 103,
Luo, S. M., Schatten, H., and Sun, Q. Y. (2013). Sperm mitochondria in reproduction:
140–150. doi:10.1016/j.bcp.2016.01.019
good or bad and where do they go? J. Genet. Genomics 40, 549–556. doi:10.1016/j.jgg.
2013.08.004 Sallmyr, A., Fan, J., and Rassool, F. V. (2008). Genomic instability in myeloid
malignancies: increased reactive oxygen species (ROS), DNA double strand breaks
Maehly, A. C., and Chance, B. (1954). The assay of catalases and peroxidases. Methods
(DSBs) and error-prone repair. Cancer Lett. 270, 1–9. doi:10.1016/j.canlet.2008.03.036
Biochem. Anal. 1, 357–424. doi:10.1002/9780470110171.ch14
Salmon, A. B., Richardson, A., and Perez, V. I. (2010). Update on the oxidative stress
Mahmoud, A. M., Goemaere, S., El-Garem, Y., Van Pottelbergh, I., Comhaire, F. H.,
theory of aging: does oxidative stress play a role in aging or healthy aging? Free Radic.
and Kaufman, J. M. (2003). Testicular volume in relation to hormonal indices of
Biol. Med. 48, 642–655. doi:10.1016/j.freeradbiomed.2009.12.015
gonadal function in community-dwelling elderly men. J. Clin. Endocrinol. Metab. 88,
179–184. doi:10.1210/jc.2002-020408 Sasaki, H., Hamatani, T., Kamijo, S., Iwai, M., Kobanawa, M., Ogawa, S., et al. (2019).
Impact of oxidative stress on age-associated decline in oocyte developmental
Mansouri Torghabeh, F., Rostamzadeh, P., Davoudi, S., Keivan, M., and Shokri-Asl,
competence. Front. Endocrinol. (Lausanne) 10, 811. doi:10.3389/fendo.2019.00811
V. (2022). Effects of Rosmarinus officinalis on orchitis following spermatic cord torsion-
detorsion in male mice with emphasis on anti-inflammatory and antioxidant properties. Sathananthan, A. H., and Trounson, A. O. (2000). Mitochondrial morphology during
Andrologia 54, e14252. doi:10.1111/and.14252 preimplantational human embryogenesis. Hum. Reprod. 15 (Suppl. 2), 148–159. doi:10.
1093/humrep/15.suppl_2.148
Manzo, S., Buono, S., and Cremisini, C. (2010). Cadmium, lead and their mixtures
with copper: Paracentrotus lividus embryotoxicity assessment, prediction, and offspring Sato, M., Sato, K., Tomura, K., Kosako, H., and Sato, K. (2018). The autophagy
quality evaluation. Ecotoxicology 19, 1209–1223. doi:10.1007/s10646-010-0506-z receptor ALLO-1 and the IKKE-1 kinase control clearance of paternal mitochondria in
Caenorhabditis elegans. Nat. Cell. Biol. 20, 81–91. doi:10.1038/s41556-017-0008-9
Marchetti, C., Obert, G., Deffosez, A., Formstecher, P., and Marchetti, P. (2002). Study
of mitochondrial membrane potential, reactive oxygen species, DNA fragmentation and Schatten, H., Prather, R. S., and Sun, Q. Y. (2005). The significance of mitochondria
cell viability by flow cytometry in human sperm. Hum. Reprod. 17, 1257–1265. doi:10. for embryo development in cloned farm animals. Mitochondrion 5, 303–321. doi:10.
1093/humrep/17.5.1257 1016/j.mito.2005.05.003
Marques-Da-Silva, C., Chaves, M. M., Castro, N. G., Coutinho-Silva, R., and Schatten, H., and Sun, Q. Y. (2015). Centrosome and microtubule functions and
Guimaraes, M. Z. (2011). Colchicine inhibits cationic dye uptake induced by ATP dysfunctions in meiosis: implications for age-related infertility and developmental
in P2X2 and P2X7 receptor-expressing cells: implications for its therapeutic action. Br. disorders. Reprod. Fertil. Dev. 27, 934–943. doi:10.1071/RD14493
J. Pharmacol. 163, 912–926. doi:10.1111/j.1476-5381.2011.01254.x
Secomandi, L., Borghesan, M., Velarde, M., and Demaria, M. (2022). The role of
Matzuk, M. M., Dionne, L., Guo, Q., Kumar, T. R., and Lebovitz, R. M. (1998). cellular senescence in female reproductive aging and the potential for
Ovarian function in superoxide dismutase 1 and 2 knockout mice. Endocrinology 139, senotherapeutic interventions. Hum. Reprod. Update 28, 172–189. doi:10.1093/
4008–4011. doi:10.1210/endo.139.9.6289 humupd/dmab038
Min, K. J., Lee, C. K., and Park, H. N. (2012). The lifespan of Korean eunuchs. Curr. Seli, E., Wang, T., and Horvath, T. L. (2019). Mitochondrial unfolded protein
Biol. 22, R792–R793. doi:10.1016/j.cub.2012.06.036 response: a stress response with implications for fertility and reproductive aging.
Fertil. Steril. 111, 197–204. doi:10.1016/j.fertnstert.2018.11.048
Mogessie, B. (2020). Advances and surprises in a decade of oocyte meiosis research.
Essays Biochem. 64, 263–275. doi:10.1042/EBC20190068 Shen, G., Zhou, L., Liu, W., Cui, Y., Xie, W., Chen, H., et al. (2017). Di(2-ethylhexyl)
phthalate alters the synthesis and β-oxidation of fatty acids and hinders ATP supply in
Murakoshi, Y., Sueoka, K., Takahashi, K., Sato, S., Sakurai, T., Tajima, H., et al. (2013).
mouse testes via UPLC-Q-exactive orbitrap MS-based metabonomics study. J. Agric.
Embryo developmental capability and pregnancy outcome are related to the
Food Chem. 65, 5056–5063. doi:10.1021/acs.jafc.7b01015
mitochondrial DNA copy number and ooplasmic volume. J. Assist. Reprod. Genet.
30, 1367–1375. doi:10.1007/s10815-013-0062-6 Shen, Q., Liu, Y., Li, H., and Zhang, L. (2021). Effect of mitophagy in oocytes and
granulosa cells on oocyte quality†. Biol. Reprod. 104, 294–304. doi:10.1093/biolre/
Murphy, C. T. (2023). Aging research: a field grows up. PLoS Biol. 21, e3002132.
ioaa194
doi:10.1371/journal.pbio.3002132
Sherratt, H. S. (1991). Mitochondria: structure and function. Rev. Neurol. Paris. 147,
Nakamura, J., Kamiya, H., Haneda, M., Inagaki, N., Tanizawa, Y., Araki, E., et al.
417–430.
(2017). Causes of death in Japanese patients with diabetes based on the results of a
survey of 45,708 cases during 2001-2010: report of the Committee on Causes of Death in Shi, S., Geng, Z., Yu, X., Hu, B., Liu, L., Chi, Z., et al. (2023). Salidroside
Diabetes Mellitus. J. Diabetes Investig. 8, 397–410. doi:10.1111/jdi.12645 supplementation affects in vitro maturation and preimplantation embryonic
development by promoting meiotic resumption. Genes. (Basel) 14, 1729. doi:10.
Noh, S. E., Lee, S. J., Lee, T. G., Park, K. S., and Kim, J. H. (2023). Inhibition of cellular
3390/genes14091729
senescence hallmarks by mitochondrial transplantation in senescence-induced ARPE-
19 cells. Neurobiol. Aging 121, 157–165. doi:10.1016/j.neurobiolaging.2022.11.003 Shuster, L. T., Rhodes, D. J., Gostout, B. S., Grossardt, B. R., and Rocca, W. A. (2010).
Premature menopause or early menopause: long-term health consequences. Maturitas
O’flaherty, C. (2019). Orchestrating the antioxidant defenses in the epididymis.
65, 161–166. doi:10.1016/j.maturitas.2009.08.003
Andrology 7, 662–668. doi:10.1111/andr.12630
Sinha, A., and Rae, R. (2014). A functional genomic screen for evolutionarily
Ozatik, F. Y., Ozatik, O., Yigitaslan, S., Kaygisiz, B., and Erol, K. (2020). Do resveratrol
conserved genes required for lifespan and immunity in germline-deficient C.
and dehydroepiandrosterone increase diminished ovarian reserve? Eurasian J. Med. 52,
elegans. PLoS One 9, e101970. doi:10.1371/journal.pone.0101970
6–11. doi:10.5152/eurasianjmed.2019.19044
Srirattana, K., and St John, J. C. (2018). Additional mitochondrial DNA influences the
Papas, M., Arroyo, L., Bassols, A., Catalan, J., Bonilla-Correal, S., Gacem, S., et al.
interactions between the nuclear and mitochondrial genomes in a bovine embryo model
(2019). Activities of antioxidant seminal plasma enzymes (SOD, CAT, GPX and GSR)
of nuclear transfer. Sci. Rep. 8, 7246. doi:10.1038/s41598-018-25516-3

Frontiers in Cell and Developmental Biology 11 frontiersin.org


Song et al. 10.3389/fcell.2024.1347286

Stone, B. A., Alex, A., Werlin, L. B., and Marrs, R. P. (2013). Age thresholds for Wassarman, P. M., Albertini, D. F., Josefowicz, W. J., and Letourneau, G. E. (1976).
changes in semen parameters in men. Fertil. Steril. 100, 952–958. doi:10.1016/j. Cytochalasin B-induced pseudo-cleavage of mouse oocytes in vitro: asymmetric
fertnstert.2013.05.046 localization of mitochondria and microvilli associated with a stage-specific response.
J. Cell. Sci. 21, 523–535. doi:10.1242/jcs.21.3.523
Suen, D. F., Norris, K. L., and Youle, R. J. (2008). Mitochondrial dynamics and
apoptosis. Genes. Dev. 22, 1577–1590. doi:10.1101/gad.1658508 Wdowiak, A., Brzozowski, I., and Bojar, I. (2015). Superoxide dismutase and
glutathione peroxidase activity in pregnancy complicated by diabetes. Ann. Agric.
Sutovsky, P., Moreno, R. D., Ramalho-Santos, J., Dominko, T., Simerly, C., and
Environ. Med. 22, 297–300. doi:10.5604/12321966.1152083
Schatten, G. (1999). Ubiquitin tag for sperm mitochondria. Nature 402, 371–372.
doi:10.1038/46466 Wei, Y., Chiang, W. C., Sumpter, R., Mishra, P., and Levine, B. (2017). Prohibitin 2 is
an inner mitochondrial membrane mitophagy receptor. Cell. 168, 224–238. doi:10.1016/
Takeuchi, T., Neri, Q. V., Katagiri, Y., Rosenwaks, Z., and Palermo, G. D. (2005).
j.cell.2016.11.042
Effect of treating induced mitochondrial damage on embryonic development and
epigenesis. Biol. Reprod. 72, 584–592. doi:10.1095/biolreprod.104.032391 Weng, Y., Zhang, Y., Wang, D., Wang, R., Xiang, Z., Shen, S., et al. (2023). Exercise-
induced irisin improves follicular dysfunction by inhibiting IRE1α-TXNIP/ROS-
Tan, J. X., and Finkel, T. (2020). Mitochondria as intracellular signaling platforms in
NLRP3 pathway in PCOS. J. Ovarian Res. 16, 151. doi:10.1186/s13048-023-01242-x
health and disease. J. Cell. Biol. 219, e202002179. doi:10.1083/jcb.202002179
Wu, M., Ma, L., Xue, L., Ye, W., Lu, Z., Li, X., et al. (2019). Resveratrol alleviates
Tarin, J. J. (1995). Aetiology of age-associated aneuploidy: a mechanism based on the ’free
chemotherapy-induced oogonial stem cell apoptosis and ovarian aging in mice. Aging
radical theory of ageing. Hum. Reprod. 10, 1563–1565. doi:10.1093/humrep/10.6.1563
(Albany NY) 11, 1030–1044. doi:10.18632/aging.101808
Terman, A., Gustafsson, B., and Brunk, U. T. (2007). Autophagy, organelles and
Wu, Y. W., Li, S., Zheng, W., Li, Y. C., Chen, L., Zhou, Y., et al. (2022). Dynamic
ageing. J. Pathol. 211, 134–143. doi:10.1002/path.2094
mRNA degradome analyses indicate a role of histone H3K4 trimethylation in
Thornton, J. (2019). WHO report shows that women outlive men worldwide. Bmj association with meiosis-coupled mRNA decay in oocyte aging. Nat. Commun. 13,
365, l1631. doi:10.1136/bmj.l1631 3191. doi:10.1038/s41467-022-30928-x
Thouas, G. A., Trounson, A. O., and Jones, G. M. (2006). Developmental effects of Wyman, A., Pinto, A. B., Sheridan, R., and Moley, K. H. (2008). One-cell zygote
sublethal mitochondrial injury in mouse oocytes. Biol. Reprod. 74, 969–977. doi:10. transfer from diabetic to nondiabetic mouse results in congenital malformations and
1095/biolreprod.105.048611 growth retardation in offspring. Endocrinology 149, 466–469. doi:10.1210/en.2007-1273
Tilly, J. L., and Sinclair, D. A. (2013). Germline energetics, aging, and female Xu, W., Sun, T., Wang, J., Wang, T., Wang, S., Liu, J., et al. (2022). GPX4 alleviates
infertility. Cell. Metab. 17, 838–850. doi:10.1016/j.cmet.2013.05.007 diabetes mellitus-induced erectile dysfunction by inhibiting ferroptosis. Antioxidants
(Basel) 11, 1896. doi:10.3390/antiox11101896
Udagawa, O., Ishihara, T., Maeda, M., Matsunaga, Y., Tsukamoto, S., Kawano, N.,
et al. (2014). Mitochondrial fission factor Drp1 maintains oocyte quality via dynamic Yang, L., Tan, G. Y., Fu, Y. Q., Feng, J. H., and Zhang, M. H. (2010). Effects of acute heat
rearrangement of multiple organelles. Curr. Biol. 24, 2451–2458. doi:10.1016/j.cub.2014. stress and subsequent stress removal on function of hepatic mitochondrial respiration, ROS
08.060 production and lipid peroxidation in broiler chickens. Comp. Biochem. Physiol. C Toxicol.
Pharmacol. 151, 204–208. doi:10.1016/j.cbpc.2009.10.010
Vadakedath, S., Kandi, V., Ca, J., Vijayan, S., Achyut, K. C., Uppuluri, S., et al. (2023).
Mitochondrial deoxyribonucleic acid (mtDNA), maternal inheritance, and their role in Yang, Q., Cong, L., Wang, Y., Luo, X., Li, H., Wang, H., et al. (2020). Increasing
the development of cancers: a scoping review. Cureus 15, e39812. doi:10.7759/cureus. ovarian NAD(+) levels improve mitochondrial functions and reverse ovarian aging.
39812 Free Radic. Biol. Med. 156, 1–10. doi:10.1016/j.freeradbiomed.2020.05.003
Vashisht, A., and Gahlay, G. K. (2023). Understanding seminal plasma in male Yang, Q., Xi, Q., Wang, M., Long, R., Hu, J., Li, Z., et al. (2022). Rapamycin improves the
infertility: emerging markers and their implications. Andrology. doi:10.1111/andr.13563 quality and developmental competence of mice oocytes by promoting DNA damage repair
during in vitro maturation. Reprod. Biol. Endocrinol. 20, 67. doi:10.1186/s12958-022-00943-0
Venkatesh, S., Deecaraman, M., Kumar, R., Shamsi, M. B., and Dada, R. (2009). Role
of reactive oxygen species in the pathogenesis of mitochondrial DNA (mtDNA) Yao, X., Zhu, J., Li, L., Yang, B., Chen, B., Bao, E., et al. (2023). Hsp90 protected
mutations in male infertility. Indian J. Med. Res. 129, 127–137. chicken primary myocardial cells from heat-stress injury by inhibiting oxidative stress
and calcium overload in mitochondria. Biochem. Pharmacol. 209, 115434. doi:10.1016/j.
Wai, T., Ao, A., Zhang, X., Cyr, D., Dufort, D., and Shoubridge, E. A. (2010). The role
bcp.2023.115434
of mitochondrial DNA copy number in mammalian fertility. Biol. Reprod. 83, 52–62.
doi:10.1095/biolreprod.109.080887 Yin, T., Yue, X., Li, Q., Zhou, X., Dong, R., Chen, J., et al. (2023). The association
between the levels of oxidative stress indicators (MDA, SOD, and GSH) in seminal
Wai, T., and Langer, T. (2016). Mitochondrial dynamics and metabolic regulation.
plasma and the risk of idiopathic oligo-asthenotera-tozoospermia: does Cu or Se level
Trends Endocrinol. Metab. 27, 105–117. doi:10.1016/j.tem.2015.12.001
alter the association? Biol. Trace Elem. Res. doi:10.1007/s12011-023-03888-6
Wang, F., Yin, Y., Nie, X., Zou, Y., Tong, X., Tong, Y., et al. (2023a). Protocatechuic
Yu, Y., Dumollard, R., Rossbach, A., Lai, F. A., and Swann, K. (2010). Redistribution
acid alleviates polycystic ovary syndrome symptoms in mice by PI3K signaling in
of mitochondria leads to bursts of ATP production during spontaneous mouse oocyte
granulosa cells to relieve ROS pressure and apoptosis. Gynecol. Endocrinol. 39, 2228917.
maturation. J. Cell. Physiol. 224, 672–680. doi:10.1002/jcp.22171
doi:10.1080/09513590.2023.2228917
Zhang, M., Bener, M. B., Jiang, Z., Wang, T., Esencan, E., Scott, R., Iii, et al. (2019).
Wang, J., Bai, L., Li, J., Sun, C., Zhao, J., Cui, C., et al. (2009a). Proteomic analysis of
Mitofusin 1 is required for female fertility and to maintain ovarian follicular reserve.
mitochondria reveals a metabolic switch from fatty acid oxidation to glycolysis in the
Cell. Death Dis. 10, 560. doi:10.1038/s41419-019-1799-3
failing heart. Sci. China C Life Sci. 52, 1003–1010. doi:10.1007/s11427-009-0140-2
Zhang, Q., Hao, J. X., Liu, B. W., Ouyang, Y. C., Guo, J. N., Dong, M. Z., et al. (2023).
Wang, J., Jia, R., Gong, H., Celi, P., Zhuo, Y., Ding, X., et al. (2021a). The effect of
Supplementation of mitochondria from endometrial mesenchymal stem cells improves
oxidative stress on the chicken ovary: involvement of microbiota and melatonin
oocyte quality in aged mice. Cell. Prolif. 56, e13372. doi:10.1111/cpr.13372
interventions. Antioxidants (Basel) 10, 1422. doi:10.3390/antiox10091422
Zhang, R., Wang, Y., Ye, K., Picard, M., and Gu, Z. (2017). Independent impacts of
Wang, L. Y., Wang, D. H., Zou, X. Y., and Xu, C. M. (2009b). Mitochondrial functions
aging on mitochondrial DNA quantity and quality in humans. BMC Genomics 18, 890.
on oocytes and preimplantation embryos. J. Zhejiang Univ. Sci. B 10, 483–492. doi:10.
doi:10.1186/s12864-017-4287-0
1631/jzus.B0820379
Zhang, X., Wu, X. Q., Lu, S., Guo, Y. L., and Ma, X. (2006). Deficit of mitochondria-
Wang, X., Sharma, R. K., Gupta, A., George, V., Thomas, A. J., Falcone, T., et al.
derived ATP during oxidative stress impairs mouse MII oocyte spindles. Cell. Res. 16,
(2003). Alterations in mitochondria membrane potential and oxidative stress in infertile
841–850. doi:10.1038/sj.cr.7310095
men: a prospective observational study. Fertil. Steril. 80 (2), 844–850. doi:10.1016/
s0015-0282(03)00983-x Zhang, Y., and Hood, W. R. (2016). Current versus future reproduction and longevity:
a re-evaluation of predictions and mechanisms. J. Exp. Biol. 219, 3177–3189. doi:10.
Wang, X., Wen, Y., Zhang, J., Swanson, G., Guo, S., Cao, C., et al. (2021b).
1242/jeb.132183
MFN2 interacts with nuage-associated proteins and is essential for male germ cell
development by controlling mRNA fate during spermatogenesis. Development 148, Zhang, Y., Wang, C., Jin, Y., Yang, Q., Meng, Q., Liu, Q., et al. (2018). Activating the
dev196295. doi:10.1242/dev.196295 PGC-1α/TERT pathway by catalpol ameliorates atherosclerosis via modulating ROS
production, DNA damage, and telomere function: implications on mitochondria and
Wang, Y., Chen, X., Baker, J. S., Davison, G. W., Xu, S., Zhou, Y., et al. (2023b).
telomere link. Oxid. Med. Cell. Longev. 2018, 2876350. doi:10.1155/2018/2876350
Astaxanthin promotes mitochondrial biogenesis and antioxidant capacity in chronic
high-intensity interval training. Eur. J. Nutr. 62, 1453–1466. doi:10.1007/s00394-023- Zhao, H. C., Ding, T., Ren, Y., Li, T. J., Li, R., Fan, Y., et al. (2016). Role of Sirt3 in
03083-2 mitochondrial biogenesis and developmental competence of human in vitro matured
oocytes. Hum. Reprod. 31, 607–622. doi:10.1093/humrep/dev345
Wang, Y., Xu, J., Fan, Z., Zhou, X., Wang, Z., and Zhang, H. (2023c). Unlocking the
antioxidant potential of white tea and osmanthus flower: a novel polyphenol liquid Zhou, C., Zhang, X., Chen, Y., Liu, X., Sun, Y., and Xiong, B. (2019). Glutathione
preparation and its impact on km mice and their offspring. Foods 12, 4041. doi:10.3390/ alleviates the cadmium exposure-caused porcine oocyte meiotic defects via eliminating
foods12214041 the excessive ROS. Environ. Pollut. 255, 113194. doi:10.1016/j.envpol.2019.113194
Wang, Z. A., Huang, J., and Kalderon, D. (2012). Drosophila follicle stem cells are Zhou, L. H., Zou, H., Hao, J. Y., Huang, Y., Zhang, J. N., Xu, X. H., et al. (2023).
regulated by proliferation and niche adhesion as well as mitochondria and ROS. Nat. Metformin inhibits ovarian granular cell pyroptosis through the miR-670-3p/NOX2/
Commun. 3, 769. doi:10.1038/ncomms1765 ROS pathway. Aging (Albany NY) 15, 4429–4443. doi:10.18632/aging.204745

Frontiers in Cell and Developmental Biology 12 frontiersin.org

You might also like