Next Article in Journal
The Effect of Heat Stress During the Insemination Period on the Conception Outcomes of Dairy Cows
Previous Article in Journal
Adaptation Strategy of the Planula Strobilation in Moon Jelly, Aurelia coerulea to Acidic Environments in Terms of Statolith Formation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitochondrial Quality Control in Bovine Oocyte Maturation: Mechanisms, Challenges, and Prospects for Enhancing Reproductive Efficiency

Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
*
Author to whom correspondence should be addressed.
Animals 2025, 15(13), 2000; https://doi.org/10.3390/ani15132000
Submission received: 14 May 2025 / Revised: 19 June 2025 / Accepted: 3 July 2025 / Published: 7 July 2025
(This article belongs to the Section Animal Reproduction)

Simple Summary

The past decades have witnessed significant advancements in live-cell suborganelle imaging technologies and multi-omics approaches, providing powerful analytical tools to investigate mitochondrial functional dynamics during bovine oocyte maturation. Nevertheless, persistent challenges in mitochondrial dysfunction and compromised oocyte maturation quality continue to constrain mammalian reproductive efficiency and biological research progress. Emerging evidence suggests that dysregulation of mitochondrial quality control (MQC) networks may underlie these abnormalities by disrupting critical physiological processes, including energy metabolism, meiotic regulation, and intracellular signaling pathways. This review elucidates the mechanistic regulation of bovine oocyte maturation through MQC pathways, offering novel perspectives for improving reproductive efficiency by targeting mitochondrial function.

Abstract

Oocyte maturation represents a fundamental biological process in bovine reproduction, establishing the physiological basis for fertilization and early embryonic development while critically determining the propagation of improved varieties and breeding efficiency. The roles of MQC in reproduction have gained substantial scientific attention. The proper maturation of oocytes fundamentally depends on adequate mitochondrial functionality. However, the intrinsic regulatory mechanisms governing MQC during bovine oocyte maturation remain incompletely characterized. Here, we discuss the most recent progress on the molecular mechanisms and roles of mitochondrial fission/fusion, biogenesis, and mitophagy in MQC. Building upon the mechanistic foundations of MQC in bovine oocyte maturation, this review identifies key mitochondrial-targeted supplements with potential applications in enhancing oocyte quality. Furthermore, we evaluate epigenetic influences on mitochondrial regulatory networks through mitochondrial–nuclear communication. Finally, we discuss the challenges in elucidating mitochondrial quality control mechanisms during oocyte maturation and propose corresponding strategies to address these obstacles. Integrating mechanistic insights, this review proposes strategies to enhance in vitro culture systems and identify oocyte quality markers, providing valuable insights for optimizing in vitro production (IVP) of bovine embryos and enhancing reproductive efficiency.

1. Introduction

Oocyte quality stands as a pivotal determinant of female reproductive success in cattle, critically governing fertilization, embryonic development, and postnatal growth trajectories [1,2]. During maturation, oocytes undergo sequential biological milestones, including cytoplasmic maturation, chromosomal recombination, and meiotic progression, to yield fully functional gametes. Mitochondria serve as essential organelles in oocytes through multiple mechanisms. Primarily, they sustain energy demands during oocyte maturation via continuous ATP production through integrated metabolic pathways, particularly oxidative phosphorylation coupled with the tricarboxylic acid cycle [3]. Furthermore, mitochondria physically and functionally interact with the endoplasmic reticulum to modulate intracellular calcium fluxes. These calcium ions act as critical signaling mediators that coordinate meiotic resumption and cell cycle progression [3]. Concurrently, mitochondrial ATP generation inherently produces reactive oxygen species (ROS), where excessive accumulation may induce oxidative damage through protein denaturation and lipid peroxidation [4]. Mitochondria also regulate cellular fate through metabolic intermediates that influence signaling cascades, exemplified by cytochrome C release initiating apoptotic pathways [4]. Notably, tricarboxylic acid cycle metabolites directly mediate epigenetic modifications: acetyl-CoA facilitates histone acetylation while α-ketoglutarate modulates DNA demethylation through enzymatic regulation.
Mitochondrial quality control (MQC) constitutes a central regulatory axis governing oocyte maturation, coordinating ATP biogenesis, selective elimination of damaged organelles, preservation of mtDNA integrity, and dynamic mitochondrial reorganization. Earlier studies focused on nutritional influences or individual MQC components. However, recent research demonstrates that mitochondrial biogenesis, dynamics (particularly the fusion/fission balance), and selective autophagy (mitophagy) work synergistically during bovine oocyte maturation. Notably, follicular development synchronizes morphological and quantitative adaptations in oocytes and granulosa cells (GCs), with mitochondrial abundance, spatial distribution, and functional integrity directly dictating developmental outcomes [5,6]. Dysregulated mitochondrial dynamics and compromised mitophagy not only impair ovarian architecture but also exacerbate oxidative stress in GCs, while emerging evidence links these perturbations to diminished oocyte quality and broader reproductive health consequences [7,8]. This synthesis elucidates the molecular interplay of MQC mechanisms governing bovine oocyte maturation, proposing targeted strategies to enhance in vitro maturation systems, mitigate age-related fertility decline, and optimize breeding protocols for improved livestock reproductive efficiency and herd sustainability. This review systematically discussed the impact of MQC mechanisms on bovine oocyte maturation through molecular regulatory networks, synthesizes key regulatory pathways identified in current research, and proposes potential strategies to enhance mitochondrial health for oocytes.

2. Oocyte Maturation

Oocytes maturation progresses through three critical stages: germinal vesicle breakdown (GVBD), metaphase I (MI), and metaphase II (MII). The initial phase involves the growth and proliferation of primordial oocytes, during which they gradually acquire the morphological and structural features of mature gametes. This process encompasses two parallel mechanisms: nuclear maturation (involving chromosomal segregation) and cytoplasmic maturation (marked by organelle reorganization and energy accumulation). Studies have identified multiple signaling pathways, including the mTOR [9,10] and AMPK pathways [11,12], as well as translational control mechanisms, that regulate oocyte development. Embryos derived from oocytes with inhibited mTOR signaling during ovulation exhibit insufficient energy supply and reduced developmental potential [13]. Notably, in vitro maturation studies of bovine oocytes demonstrate that mTOR inhibition enhances energy metabolism, highlighting its critical role in balancing oocyte bioenergetics. Current evidence confirms the essential role of mTOR in sustaining mitochondrial oxidative capacity. mTOR inhibition rapidly disrupts carbon utilization and mitochondrial metabolism in leukemia cell lines. Leukemic cells treated with rapamycin exhibit impaired mitochondrial function, resulting in preferential reliance on aerobic glycolysis over mitochondrial respiration for energy production [14]. Investigations demonstrate that mTOR exerts metabolic control through a Bcl-xl-VDAC1 complex localized at the mitochondrial outer membrane. In skeletal muscle systems, rapamycin suppresses the expression of mitochondrial transcriptional regulators (PGC-1α, ERRα, NRF), consequently reducing mitochondrial gene expression and oxygen consumption. Pharmacological mTOR inhibitors induce mitochondrial hyperfusion, aberrant branching, and disrupted fission processes via mTORC1 axis decoupling, ultimately triggering apoptotic cell death [14,15,16]. Specifically, bovine oocytes treated with Juglone and matured in vitro showed downregulation of the PI3K/AKT/mTOR pathway and its downstream signals [17]. This was manifested by autophagy induction with significantly increased levels of LC3B and beclin-1; increased DNA damage with overexpression of DNA damage-specific markers (8-Oxoguanine; 8-OxoG); significant downregulation of glycolysis-specific genes (PFK1 and GLUT1); and significantly reduced expression of ATP synthesis genes (ATPase8, ATP5F1B) in oocytes [18]. Normally, the translation initiation factor eIF4E (cap-binding protein) undergoes progressive phosphorylation during meiotic maturation of bovine oocytes. At the GVBD-mid I transition, eIF4E phosphorylation coincides with a significant increase in protein synthesis, whereas at mid II, oocyte protein synthesis declines to levels comparable to GV oocytes despite full eIF4E phosphorylation [19]. eIF4E phosphorylation timing strongly correlates with MAP kinase activation. Furthermore, the eIF4E-specific repressor 4E-BP1 forms a complex with eIF4E; enhanced eIF4E/4E-BP1 complex formation in mid-II oocytes coincides with reduced translation rates at this stage, suggesting 4E-BP1 involvement in inhibiting cap-dependent translation in mid-II bovine oocytes [20]. Interestingly, the study found that 4E-BP1 becomes progressively phosphorylated from the GVBD stage to the MII stage, while the mTOR inhibitor Torin2 irreversibly arrests bovine oocyte maturation at the MI stage. Torin2 treatment inhibits translation by enhancing 4E-BP1 binding to eIF4E [21], thereby suppressing 4E-BP1 phosphorylation at Thr37/46 in a reversible manner. However, no significant differences were observed in mTOR expression levels or phosphorylation between IVM and Torin2-treated groups. Collectively, the roles of mTOR and eIF4E in bovine oocyte maturation require further investigation [22].
Chromosomal recombination and meiosis are central to nuclear maturation, wherein crossovers’ formation and synaptonemal complex assembly ensure accurate chromosomal segregation through physical chiasma stabilization. Mitochondria cooperate with the meiotic spindle to achieve asymmetric division. The presence of formin2 (FMN2) around oocytes induces the aggregation of short actin bundles derived from endoplasmic reticulum vesicles at the spindle periphery, which subsequently drives mitochondria associated with the spindle, promoting their redistribution and facilitating spindle migration [23,24]. Recent evidence links mitochondrial activity fluctuations to meiotic competence: developmentally competent oocytes exhibit two distinct peaks of mitochondrial activation (prior to meiotic resumption and before maturation completion), while suboptimal oocytes show only one activation event preceding maturation [25,26]. Overexpression of mitochondrial fusion protein MFN2 increases mitochondrial density near the spindle in mouse oocytes without significantly altering mitochondrial membrane potential (MMP) or ROS production [27]. This suggests that MFN2 influences nuclear maturation of oocyte through the mitochondrial pathway. Paradoxically, excessive mitochondrial accumulation around the spindle inhibits spindle movement and chromosomal separation, causing metaphase I arrest [27]. Mitochondrial Rho-GTPase 1 (MIRO1) regulates mitochondrial transport and mitophagy during meiosis. MIRO1 deficiency disrupts meiotic resumption and polar body extrusion in mouse and pig oocytes. Mechanistically, MIRO1 interacts with microtubule-organizing center (MTOC)-associated motor proteins to regulate Aurora Kinase A (Aurora A) and kinesin KIF11, thereby promoting spindle assembly [28]. Furthermore, MIRO1 coordinates with dynamin-related protein 1 (DRP1), Parkin, and lysosome-associated membrane glycoprotein 2 (LAMP2) to regulate mitochondrial dynamics and quality control during oocyte maturation [28,29].
The cytoplasmic maturation of oocytes is a prerequisite for normal fertilization, with mitochondrial redistribution and cortical granule maturation serving as key indicators. Mitochondria primarily generate ATP via oxidative phosphorylation (OXPHOS) through an electron transport chain (ETC) comprising five complexes (I–V) on the inner mitochondrial membrane [30]. The proton gradients established by respiratory chain complexes I, III, and IV power ATP synthase-catalyzed phosphorylation, whereas complexes I and III serve as primary sites for superoxide generation through electron leakage at flavin mononucleotide (FMN) clusters and Q-cycle intermediates, respectively [31]. This ROS overproduction triggers mitochondrial membrane potential collapse and mtDNA mutations via oxidative chain reactions [32,33]. Mitochondrial activation and spatial reorganization directly influence cytoplasmic maturation, with mitochondrial integrity determining oocyte developmental competence and fertilization success. In bovine oocytes, active mitochondria transition from cortical localization at the GVBD stage to cytoplasmic dispersion by MII. This redistribution is absent in developmentally incompetent oocytes, where mitochondria remain peripherally restricted [34,35].
Emerging evidence demonstrates that epigenetic remodeling accompanies oocyte maturation. Histone H4 acetylation and dynamic expression of histone deacetylases (HDACs) are observed during bovine oocyte maturation [36]. Mitochondrial metabolites act as enzymatic cofactors or inhibitors. For instance, DNA methyltransferases transfer methyl groups from S-adenosylmethionine (SAM) to cytosine residues, forming 5-methylcytosine (5mC), which can be passively diluted or actively removed by ten-eleven translocation (TET) enzymes in an α-ketoglutarate (α-KG)-dependent manner [37]. Metabolic interventions reducing the α-KG/succinate ratio increase global H3K27 and DNA methylation in mouse embryonic stem cells, while α-KG supplementation rescues embryos with defective epigenetic reprogramming [38]. These findings collectively demonstrate that oocyte maturation integrates cellular growth, nuclear–cytoplasmic coordination, and mitochondrial metabolic adaptation, with profound implications for improving oocyte quality in reproductive biotechnology (Figure 1).
Most experiments did not specify oxygen conditions, indicating they were conducted under ambient atmospheric oxygen. Additionally, studies revealed an interaction between oxygen tension and oocyte density that increases ROS production under specific combinations, subsequently affecting bovine oocyte in vitro maturation quality [39]. This may be attributed to the elevated oxidative stress induced by reactive oxygen species (ROS) under hyperoxic conditions, which in turn triggers abnormalities in mitochondrial autophagy regulation, ultimately impairing the maturation quality of oocytes. Future studies should further elucidate the mechanism underlying the relationship between distinct oxygen concentrations and bovine oocyte maturation.

3. Mitochondrial Quality Control

MQC is an integrated regulatory network responsible for monitoring mitochondrial quality, which is crucial for maintaining mitochondrial homeostasis and functionality in cells. MQC orchestrates mitochondrial homeostasis through coordinated processes, including mitochondrial biogenesis, fission and fusion, proteolysis, autophagic degradation, and selective clearance of damaged mitochondrial inner membranes. Within the mitochondrial quality control system, mitochondrial biogenesis represents a critical compensatory mechanism activated in oocytes experiencing elevated energy demands or cellular stress. This process involves continuous mtDNA replication, restoration of mtDNA integrity and electron transport chain components, coupled with the replacement of dysfunctional mitochondria with functional organelles to maintain adequate ATP production. The regulation of mitochondrial biogenesis demonstrates intricate coordination between mtDNA and nuclear genomic mechanisms [40,41]. Mitochondrial dynamics encompasses the perpetual cycle of fission and fusion events that dynamically regulate organellar morphology, size, and intracellular distribution to adapt to fluctuating energy requirements and cellular signaling demands [42,43]. Mitochondrial proteostasis, mediated through specialized proteolytic systems, constitutes an essential defense mechanism against oxidative damage, protein misfolding, and electron transport chain deficiencies. Within the mitochondrial matrix, a collaborative network of proteases, including the CLPXP complex and Lon protease homolog (LONP), orchestrates the selective degradation of aberrant proteins while regulating metabolic enzyme turnover [44]. The quality control cascade culminates in mitophagy, a selective autophagic process that ensures cellular homeostasis through targeted elimination of irreparably damaged mitochondria, thereby preserving optimal mitochondrial function and genomic integrity [45,46].
Under oxidative stress, studies demonstrate that SOD2 acetylation significantly increases mtROS production. Honokiol supplementation markedly elevates SIRT3 protein levels and restores the NaF-reduced SOD2 acetylation form in Western blotting (WB) analysis (p < 0.05) [47]. SIRT3 inhibition reduces deacetylated superoxide dismutase 2 (SOD2) levels, triggering mitochondrial ROS-induced oxidative stress-mediated apoptosis. This disrupts redox balance during oocyte maturation and causes mitochondrial damage [47,48]. Isorhamnetin activates the PI3K/Akt pathway, counteracting oxidative stress and mitochondrial dysfunction significantly (p < 0.05) to promote maturation [49].
Mitochondrial metabolites also critically influence oocyte maturation. In bovine oocytes, pyruvate conversion to acetyl-CoA is modulated by sodium dichloroacetate (DCA), which elevates H3K9ac levels, alters transcriptional activity, and potentially compromises oocyte quality [50]. Pyruvate metabolic disturbances during maturation activate β-oxidation, disrupt mitochondrial metabolism, and reduce mRNA content [50,51]. L-carnitine facilitates fatty acid β-oxidation by transporting activated fatty acids into mitochondria via carnitine palmitoyltransferase I (CPT1), generating acetyl-CoA for ATP production [52]. Supplementation with L-carnitine in IVM media improves embryo outcomes by providing essential cofactors for fatty acid utilization. Recent findings highlight its role in balancing acetyl-CoA/CoA ratios, sustaining glucose metabolism, enhancing mitochondrial energy output, and alleviating endoplasmic reticulum stress by shuttling or eliminating toxic palmitate [53]. Additionally, L-carnitine improves mitochondrial distribution uniformity in mature oocytes by scavenging ROS [54,55].
Collectively, the MQC network operates dynamically to maintain mitochondrial integrity. Supplementation with resveratrol or melatonin enhances mitochondrial activity, upregulates biogenesis and fission/fusion significantly (p < 0.05), and mitigates dysfunction in damaged mitochondria. While nutritional studies elucidate MQC mechanisms via pathways such as PGC1α/NRFs [56,57], PINK1/Parkin [58,59], cAMP-PKA [60], PI3K/AKT [61,62], and Drp1 [63], most evidence derives from murine and porcine models. The intrinsic molecular mechanisms governing MQC in bovine oocytes remain poorly characterized, warranting further investigation (Figure 2).

4. Mitochondrial Biogenesis and Its Molecular Role in Oocyte Maturation

Mitochondrial biogenesis serves as a homeostatic mechanism to sustain mitochondrial quantity by continuously replenishing damaged mitochondria with fully functional counterparts [64]. As a core determinant of mitochondrial function, this process plays a pivotal role in oocyte cytoplasmic maturation. Mitochondrial biogenesis involves a complex cascade of events, requiring coordinated activation of multiple factors under diverse biological stimuli. During meiotic resumption and cytoplasmic maturation, oocytes demand substantial ATP production, primarily supplied through mitochondrial oxidative phosphorylation. Throughout meiotic maturation, mitochondria dynamically redistribute within the oocyte, accumulating around the spindle apparatus during the first meiotic division [65]. This spatial coordination between mitochondrial distribution and meiotic spindle integrity is critical, as ATP deficiency disrupts spindle morphology and meiotic progression. Experimental evidence confirms that silencing mitochondrial-associated genes severely compromises spindle architecture and meiotic completion [65,66].
Current research focuses on key signaling pathways, notably peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a master transcriptional coactivator that drives mitochondrial biogenesis through upregulating nuclear respiratory factors (NRF1 and NRF2) and mitochondrial transcription factor A (TFAM) (Figure 3) [67,68]. This pathway enhances mitochondrial DNA replication, electron transport chain (ETC) component synthesis, and ATP generation. Mature oocytes exhibit elevated mtDNA content, underscoring the necessity for stringent regulation of mitochondrial replication and biogenesis during maturation. Notably, resveratrol depletion significantly upregulates critical biogenesis-related genes such as PGC-1α and TFAM [69,70]. In porcine oocytes, SIRT1 activation has been shown to enhance mitochondrial degradation and biogenesis, thereby improving oocyte quality. The study revealed that TFAM, PGC1α, and PINK1 expression levels were all higher in the treatment group compared to the untreated group [71,72]. Recent studies reveal that rotenone suppresses SIRT1 levels during in vitro maturation (IVM) of porcine oocytes, exacerbating mitochondrial dysfunction, increasing mitophagy, and impairing mitochondrial biogenesis, ultimately disrupting oocyte maturation [73]. Early research proposed that mitochondrial transcription factor A (TFAM) exhibited higher expression in morphologically superior oocytes regardless of follicular size, suggesting its association with developmental competence [26,74]. Subsequent investigations demonstrate that SIRT2 inhibition downregulated TFAM, perturbing mitochondrial biogenesis and dynamics during oocyte maturation [75,76]. Pre-IVM treatment with C-type natriuretic peptide (CNP) upregulated mitochondrial biogenesis-related genes, promoted mitochondrial biogenesis, and enhanced cytoplasmic maturation of oocytes [67,68]. In porcine oocytes, 10 μM of nobiletin enhanced mitochondrial biogenesis by elevating SIRT1 and PGC-1α protein levels. This intervention led to increased active mitochondrial populations, mtDNA copy number, mitochondrial membrane potential, and ATP output, thereby improving mitochondrial functionality and promoting in vitro maturation. While multiple studies in bovine oocytes and other species correlate elevated mtDNA content with enhanced developmental competence, the molecular basis underlying this association remains ambiguous [51,77,78]. This persistent knowledge gap necessitates further investigation to reconcile discrepant findings in the existing literature.
Additionally, FOXO1 and FOXO3a serve as critical signaling components in mitochondrial biogenesis. Research indicates that AMPK collaborates with SIRT1 to modulate the expression of energy metabolism-related genes in skeletal muscle. AMPK elevates cellular nicotinamide adenine dinucleotide (NAD+) levels, thereby enhancing SIRT1 activity and promoting deacetylation and functional regulation of downstream targets such as FOXO1 and FOXO3a [79]. In neuronal contexts, SIRT2 regulates mitochondrial biogenesis through deacetylation-mediated activation of transcription factors FOXO1, FOXO3a, and TFEB, along with the coactivator PGC-1α [80]. Overall, FoxO1 and FoxO3a function as suppressors of mitochondrial biogenesis through distinct molecular mechanisms. In renal tubular epithelial cells, FoxO1 exerts its inhibitory effect by blocking the formation of the CREB-CBP-P300 transcriptional complex. This disruption subsequently reduces PGC-1α expression and impairs mitochondrial biogenesis. In contrast to FoxO1, FoxO3-mediated transcriptional suppression functions through a distinct pathway independent of PGC-1 family members or NRF1. This inhibitory mechanism specifically involves targeting c-Myc, a key transcription factor that activates nuclear-encoded mitochondrial genes via direct binding to the TFAM promoter [81].
In summary, although the PGC1α/NRF1/2 axis, TFAM-mediated regulation, and FOXO1/FOXO3A signaling are well-established biomarkers for mitochondrial biogenesis, with recent advances in understanding mTOR [82,83] and p53 [84] signaling during bovine oocyte maturation, the molecular crosstalk between mitochondrial biogenesis and MQC networks remains inadequately characterized.

5. Mitochondrial Dynamics and Their Regulatory Mechanisms in Oocyte Maturation

As highly dynamic organelles, mitochondria undergo continuous fusion–fission cycles to regulate their morphology, size, and subcellular distribution. This dynamic process thereby ensures precise ATP flux regulation for cellular processes critical to oocyte maturation [85,86]. Key regulators include mitofusin 1/2 (MFN1/2) and optic atrophy 1 (OPA1) for mitochondrial fusion, while mitochondrial dynamics-related protein 1 (DRP1) governs fission (Figure 4) [87]. During oocyte transition to metaphase I (MI stage), mitochondria stage-specifically assemble into larger clusters with perinuclear polarization, maintaining spatial segregation from the spindle apparatus in a spindle apparatus-independent manner. Notably, individual and small mitochondrial clusters persist in even distribution despite reduced abundance compared to the premeiotic stage. By metaphase II (MII), mitochondrial clustering intensifies, forming larger aggregates than those in MI oocytes. Recent studies demonstrate that overexpression of mitochondrial fusion proteins induces cytoplasmic mitochondrial aggregation, disrupting spindle organization and spatiotemporal endoplasmic reticulum distribution [85]. Furthermore, oocyte-specific DRP1 knockdown impairs calcium signaling and cell–cell communication, resulting in follicular developmental defects and ovulation abnormalities [88].
Emerging evidence implicates mitochondria-associated membranes (MAMs) and mitochondria-ER contacts (MERCs) in cytoplasmic maturation deficits of oocytes from obese mice, potentially mediated by elevated Ca2+ levels, with significant differences (p < 0.05) compared to the control group [89,90]. Paradoxically, elevated perichromosomal mitochondrial Ca2+ may enhance mitochondrial energy output to support calmodulin-responsive spindle formation [91,92]. During reproductive aging in C. elegans, increased GTP-dependent mitochondrial fission near the inner membrane may drive EAT-3-mediated mitochondrial fusion, with this dynamic imbalance contributing to oocyte quality decline [93]. Sirt3 deficiency exacerbates superoxide accumulation and spindle disassembly in aged oocytes, accompanied by mitochondrial dysfunction characterized by uneven distribution, reduced membrane potential, and decreased mtDNA content. Studies reveal that compared with that of the wild type, mitochondrial membrane potential was significantly reduced in the aged group (p < 0.01), and mtDNA copy number was significantly reduced (p < 0.01) [94]. Emerging evidence highlights the critical role of optic atrophy 1 (OPA1). Defects in this inner mitochondrial membrane protein promote excessive mitochondrial fission and dysfunction [95,96]. Similarly, deoxyguanosine kinase (DGUOK) deficiency in murine oocytes leads to aberrant mitochondrial dynamics and marked reduction in mitochondrial DNA (mtDNA) copy number (p < 0.05) [97,98]. A study by further reports that deletion of mitochondrial outer membrane proteins MIGA1/2 induces cytoplasmic mitochondrial dynamics defects, potentially due to significantly elevated (p < 0.05) intra-mitochondrial ROS levels, reduced ΔΨm, and diminished mtDNA content compared to the wild type [99,100]. Additionally, hyperthermia impairs bovine oocyte embryonic development, with recent studies confirming its adverse effects on mitochondrial dynamics in porcine oocytes [101]. In mouse ovaries, Sirt3 deficiency diminishes mitochondrial respiratory chain complex activity and downregulates key proteins associated with mitochondrial fusion (OPA1, p < 0.001; MFN2, p < 0.05) and fission (DRP1, p < 0.001; FIS1, p < 0.001), ultimately compromising overall mitochondrial functionality [94]. These findings emphasize the crucial role of SIRT3 in maintaining ovarian follicle reserves and oocyte quality with aging [94,102]. Mfn2 deletion induces profound transcriptomic alterations in mouse oocytes, disrupting mitochondrial and ER homeostasis, while Mfn2-deficient oocytes exhibit mitochondrial functional impairment [103,104]. Furthermore, the knockdown of activating transcription factor 5 (ATF5) in mouse oocytes disrupts mitochondrial dynamics. ATF5 is involved in regulating vitrified oocytes through the cAMP-PKA and PI3K/AKT pathways [105,106].
In summary, mitochondria preserve their functional integrity through ongoing cycles of fission and fusion, which are closely linked to lipid metabolism and ATP production. Although current research primarily concentrates on mitochondrial dynamics regulators and their connection to oocyte maturation, the molecular interplay between mitochondrial dynamics and oocyte maturation has been insufficiently investigated.

6. Molecular Mechanisms by Which Mitophagy Regulates Oocyte Maturation

Autophagy is triggered by diverse cellular stress conditions, including nutrient deprivation, oxidative stress, pathogenic infection, or inflammatory stimuli, as extensively documented [107,108]. This vital process unfolds through three distinct yet interconnected mechanisms: chaperone-mediated autophagy, microautophagy, and macroautophagy. Mitophagy, a specialized form of macroautophagy, selectively degrades mitochondria through sequential steps involving depolarization and loss of ΔΨm in damaged mitochondria, encapsulation of mitochondria by autophagosomes to form mitophagosomes, fusion of mitophagosomes with lysosomes, and lysosomal degradation of mitochondrial components [109,110]. Mitophagy mechanisms are broadly classified into ubiquitin-dependent and ubiquitin-independent pathways. The ubiquitin-dependent pathway, exemplified by the well-characterized PINK1/Parkin cascade, involves extensive ubiquitination of mitochondrial surface proteins to trigger degradation [58,111]. In this process, ΔΨm collapse in damaged mitochondria prevents PINK1 translocation to the inner mitochondrial membrane, leading to its stabilization on the outer membrane surface [112]. PINK1 undergoes autophosphorylation and recruits/activates Parkin, an E3 ubiquitin ligase that ubiquitinates mitochondrial substrates. Subsequent binding of ubiquitinated proteins to microtubule-associated protein 1 light chain 3 (LC3)adaptors initiates autophagosome formation (Figure 5) [112], ultimately eliminating ROS-producing mitochondria to prevent oxidative DNA damage, spindle defects, and apoptosis, thereby ensuring oocyte survival during meiotic progression (GVBD to MII) and cytoplasmic maturation.
Mitophagy regulation shares similarities with mitochondrial biogenesis and dynamics. Resveratrol [113], CNP [114], SIRT1/2 [115,116], OPA1 [95,96], and rotenone collectively influence mitophagy. Resveratrol and OPA1 activate the PINK1/Parkin pathway, elevating mitophagy levels. Specifically, resveratrol upregulates PINK1, Parkin, and LC3B-II while downregulating MFN2, VDAC1, and p62 [117]. CNP ameliorates oxidative damage and suppresses excessive PINK1/Parkin-mediated mitophagy, restoring mitochondrial oxidative phosphorylation and rescuing aging-related defects in murine oocytes [114]. Melatonin co-treatment mitigates gossypol-induced damage in porcine oocytes by upregulating PINK1, Parkin, and LC3 expression while downregulating p62, a protective mechanism linked to SIRT1 activation and enhanced mitophagy [115]. Conversely, SIRT2 knockdown in ovine granulosa cells reduces oocyte maturation rates and elevates mitophagy [107,118]. Rotenone treatment in porcine oocytes significantly increases LC3 and active-caspase 3 levels, indicating that rotenone-induced mitochondrial dysfunction triggers compensatory mitophagy but ultimately promotes apoptosis [73]. Studies in porcine postovulatory aging (POA) models demonstrate that DRP1-mediated mitochondrial fission and mitophagy activation are essential for oocyte quality maintenance. β-Nicotinamide mononucleotide (NMN)-mediated activation of nicotinamide adenine dinucleotide (NAD) not only enhances oocyte quality but also promotes mitophagy in POA models, establishing NAD biosynthesis as a key regulator of DRP1-dependent mitochondrial morphology and homeostasis [119]. In Drosophila oocytes, fasting downregulates the DIP1-Clueless pathway, elevating stable intron-derived RNA sisR-1 expression. Mechanistically, sisR-1 localizes to mitochondrial aggregates, inhibits polyubiquitination of outer membrane protein Porin/VDAC1, and suppresses p62-mediated mitophagy [120,121]. Refeeding reverses fasting-induced sisR-1 elevation, restoring mitophagy, oogenesis, and oocyte quality. Activation of the PRKN-mediated mitophagy pathway coincides with meiotic arrest at metaphase I, linked to reduced RAB7 activity. Notably, microinjection of constitutively active RAB7Q67L mRNA or treatment with RAB7 activator ML098 rescues mitophagy–chromosome stability defects in CCCP-treated oocytes, highlighting RAB7 activators as potential interventions for age-related oocyte quality decline [122,123]. In rat ovaries, dehydroepiandrosterone (DHEA) attenuates significantly upregulated expression of mitophagy-related markers (Pink1, Parkin, BNIP3L, LC3, AMPK, SIRT1) and AMPK phosphorylation in diminished ovarian reserve (DOR) models, effectively suppressing AMPK-SIRT1-driven mitophagy and improving ovarian reserve [124,125]. Experimental studies demonstrate that spermidine supplementation improves oocyte quality and reproductive performance in aged female mice by enhancing mitochondrial autophagy pathways through upregulated LC3 expression [126].
The BNIP3/NIX pathway has emerged as a prominent research focus in mitochondrial quality control. As an outer mitochondrial membrane protein within the BH3-only subfamily of the BCL2 family, BNIP3L/NIX exerts dual regulatory functions in autophagy processes [127,128]. Mechanistically, BNIP3L initiates autophagosome formation by dissociating the BCL2-BECN1 complex and liberating BECN1. This autophagy-inducing activity can be pharmacologically replicated using BH3 domain mimetic small molecules. Furthermore, BNIP3L demonstrates MTORC1 inhibitory capacity to activate autophagy pathways [128]. Structural analysis reveals that BNIP3 contains evolutionarily conserved LC3-interacting region (LIR) motifs at its N-terminus, enabling direct LC3 recognition and binding to trigger mitophagy. Post-translational modifications critically regulate BNIP3L-mediated mitochondrial clearance: phosphorylation events predominantly mediate mitophagy activation, whereas ubiquitination provides an alternative pathway for selective removal of BNIP3L-tagged mitochondria [128,129].
FUNDC1 serves as a crucial mitochondrial receptor protein that mediates Parkin-independent mitophagy in mammalian cells during hypoxic stress. This process kicks off with a dynamic interplay between FUNDC1’s LC3-interacting region (LIR) domain and LC3 proteins anchored on autophagosomal membranes, orchestrating the precision-guided engulfment of damaged mitochondria [130]. Furthermore, the mitophagic activity of FUNDC1 is precisely modulated by dynamic phosphorylation events at specific serine residues (Ser13 and Ser17), where different kinase pathways exert opposing regulatory effects—phosphorylation by Unc-51-like kinase 1 (ULK1) enhances mitophagy initiation while phosphorylation by casein kinase 2 (CK2) suppresses this process [131,132,133].
Although the PINK1/Parkin pathway has been extensively characterized in mitophagy regulation, the intricate molecular crosstalk between mitophagy, mitochondrial quality control networks, and oocyte maturation remains shrouded in ambiguity, demanding rigorous mechanistic investigations to unravel these complex interdependencies.

7. Epigenetic Modifications in Mitochondrial Quality Control: Molecular Mechanisms and Functional Consequences

Most mitochondrial localization proteins are related to the transcription of nuclear-encoded genes. After the DNA in the nucleus is expressed as a protein, it is transported to the mitochondria to participate in MQC. In terms of genome methylation, studies have found that in the liver, DNA Methyltransferase 1 (DNMT1) is closely related to mitochondrial biogenesis. DNMT3B mediates the hypermethylation of the mitochondrial biogenesis regulatory factor PGC-1α gene [134]. In addition, as was found in human mesenchymal stem cells, DNMT3A is associated with methylation of the PGC-1α promoter, and methylation of its promoter region inhibits PGC-1α expression. Reduced PGC-1α expression leads to increased expression of the mitochondrial fission marker Drp1 [134], which in turn promotes the mitochondrial fission process. Increased mitochondrial fission is associated with decreased mitochondrial function [135]. In the heart, oxidative stress reduces DNMT1 activity, leading to hypomethylation of mitochondrial genes related to mitochondrial biogenesis and mitochondrial dynamics, such as PGC-1α, NRF-1, and TFAM. This imbalance ultimately leads to mitochondrial damage [136,137]. Significantly reduced methylation levels were observed in the mitochondrial DNA promoters HSP1 and HSP2 within the rat hippocampus, resulting in impaired TFAM binding and a consequent diminished capacity for mitochondrial biogenesis [138,139]. In the placenta, methylation of the MT-RNR1 region of mtDNA has been implicated in driving heightened mitophagy [140,141]. Within dopaminergic neurons, hypermethylation of the Ambra1 gene may disrupt Parkin binding, amplify LC3B expression, and ultimately exacerbate mitochondrial dysfunction and neuronal damage both in vitro and in vivo through the Am-bra1/Parkin/LC3B mitophagy cascade [142,143]. Studies in rat hearts demonstrated that DNMT1 enhances ETS1 expression through methylation of the miR-152-3p promoter region, stimulates RhoH transcriptional activation, and suppresses mitochondrial autophagy in H9c2 cells [144,145]. In addition, DNMT3A-mediated hypermethylation of the secreted frizzled-related protein 3 promoter is associated with excessive mitochondrial fission [146,147].
In the realm of RNA methylation, the RNA demethylase human AlkB homolog 5 (ALKBH5) actively diminishes significantly mitochondrial membrane potential and oxygen consumption rates, while suppressing mitochondrial fission and proliferation processes [148]. Mechanistically, ALKBH5 catalyzes the removal of N6-methyladenosine (m6A) methylation marks within the 3′ untranslated region of Drp1 transcripts, thereby downregulating Drp1 protein levels significantly and attenuating mitochondrial fission processes [134]. Furthermore, studies demonstrate that overexpression of the human mitochondrial transcription factor B1 induces pronounced hypermethylation of 12S rRNA, culminating in significant mitochondrial biogenesis abnormalities [149,150]. Research has revealed that m6A RNA methylation plays a pivotal role in regulating mitochondrial function by facilitating the translation of nuclear-encoded mitochondrial complex subunit RNAs [151]. Mettl14 knockout-induced m6A deficiency significantly reduces metabolites related to energy metabolism [152]. Tu et al. (2023) had revealed that METTL3 downregulation suppresses mitochondrial fission [153]. This process is driven by METTL3-mediated m6A methylation of the long non-coding RNA GAS5, which accelerates its degradation [153]. Intriguingly, GAS5 exhibits direct molecular interaction with Drp1, a pivotal mitochondrial fission protein, and its overexpression potently blocks Drp1-dependent mitochondrial fragmentation [154]. Furthermore, studies have demonstrated that m6A methylation at the A2212 site within OPA1’s coding sequence enhances mRNA stability, thereby promoting mitochondrial fusion [155]. Remarkably, therapeutic targeting of m6A modification through either the methyl-transferase inhibitor STM2457 or the dm6A CRISPR system produces profound inhibitory effects on mitochondrial fusion processes [156].
In the regulatory landscape of histone modification, protein arginine methyltransferase 5 (PRMT5) emerges as a crucial epigenetic modulator that catalyzes the symmetric di-methylation of arginine residues at histone H4R3, H3R8, and H2AR3 loci [134]. This enzymatic activity mechanistically establishes transcriptional repression, effectively downregulating PGC-1α expression through chromatin remodeling pathways. PRMT5 silencing or complete deletion significantly boosts PGC-1α expression and stimulates mitochondrial biogenesis [134]. Studies have demonstrated that endurance exercise training in the rat gastrocnemius muscle modulates histone methylation patterns (H3K4me3 and/or H3K27me3) within the promoter regions of the PGC-1α gene via specific epigenetic regulatory mechanisms, leading to a significant upregulation of both PGC-1α mRNA and protein expression levels (p < 0.01), which consequently enhances mitochondrial biogenesis [157,158]. Mitochondrial biogenesis is enhanced by histone H3 acetylation, in particular H3 lysine 14 acetylation (H3K14ac) [159,160]. In mice, succinate supplementation was found to increase permissive histone succinylation and H3K4me3 modification in the PGC-1α promoter, which correlated with higher PGC-1α expression [161]. In addition, SET And MYND Domain Containing 3 (SMYD3) was found to activate methylene tetrahydrofolate dehydrogenase 1-like by catalyzing the trimethylation of lysine at position 4 of histone subunit 3 (H3K4me3) [162], leading to the accumulation of the intracellular metabolites, which increases the expression of autophagy-related proteins LC3, PINK1, and Parkin and promotes mitochondrial autophagy [163].
Furthermore, mitochondrial–nuclear crosstalk profoundly regulates nuclear epigenetic modifications, mitochondrial functionality, and downstream metabolic byproducts, collectively governing oocyte maturation. Acetyl-CoA, a pivotal node in the tricarboxylic acid (TCA) cycle, is generated from mitochondrial precursors, including pyruvate, amino acids, and fatty acids. It mediates nuclear histone acetylation catalyzed by histone acetyltransferases (HATs), which neutralizes lysine residue charges to relax chromatin structure, facilitating transcription factor binding and gene regulation [164]. Studies reveal that the α-ketoglutarate dehydrogenase complex interacts with Jumonji C (JmjC)-domain-containing histone demethylases, competitively depleting α-ketoglutarate to inhibit JMJ activity, thereby modulating DNA/histone methylation levels, α-ketoglutarate concentration, and TCA cycle flux [165]. NAD+ and flavin adenine dinucleotide (FAD), pivotal coenzymes bridging the TCA cycle to electron transport via oxidative phosphorylation, play central roles in epigenetic regulation: FAD acts as an indispensable cofactor for lysine-specific demethylases, mitochondrial activity tightly regulating its bioavailability and redox balance, while NAD+ fuels the deacetylase activity of Sirtuins (SIRTs), which orchestrate histone acetylation dynamics [166]. Succinyl-CoA, synthesized from α-ketoglutarate and CoA via the α-ketoglutarate dehydrogenase complex (OGDH), is converted to succinate by succinyl-CoA synthetase. Succinate exits mitochondria through the dicarboxylate carrier SLC25A10 to regulate histone lysine succinylation [167]. Mitochondrial catabolism of tryptophan and lysine converges at α-ketoglutarate, which OGDH converts to succinyl-CoA. The FAD-dependent succinyl-CoA dehydrogenase subsequently catalyzes the conversion into an enzyme-bound intermediate, simultaneously yielding FADH2 to fuel electron transport [168]. Glutaryl-CoA is then decarboxylated into crotonyl-CoA, which cascades through successive metabolic transformations—becoming β-hydroxybutyryl-CoA, then acetoacetyl-CoA, before culminating in acetyl-CoA that integrates seamlessly into the TCA cycle [168]. Despite its low abundance, crotonyl-CoA-mediated histone lysine crotonylation functions as a positive transcriptional regulator and marks chromatin active regions [169,170].
In summary, epigenetic mechanisms emerge as a central regulatory nexus governing mitochondrial biogenesis, orchestrating dynamic structural adaptations, and finetuning quality control through mitophagy. The majority of these effects arise following epigenetic modifications in nuclear-encoded genes, which ultimately impact MQC proteins and consequently destabilize the MQC network. Intriguingly, certain epigenetic alterations directly target mtDNA or induce structural changes in MQC proteins (Figure 6). Nevertheless, the precise molecular mechanisms governing how epigenetic modifications steer the MQC network’s functionality within the realm of reproductive biology—particularly in shaping oocyte development and quality—remain profoundly enigmatic.

8. Methodological Advances in Mitochondrial Quality Control Research of Oocytes

Given that oocytes are significantly fewer in number compared to somatic cells, detecting and tracking molecular changes within MQCs poses a formidable challenge when relying on conventional methodologies. To propel this field forward, it is imperative to integrate cutting-edge multimodal approaches—such as dynamic subcellular organelle tracking via live-cell imaging, coupled with spatially resolved single-cell multi-omics (transcriptomics, metabolomics, proteomics, and epigenomics)—to holistically address persistent technological constraints. When it comes to high-resolution live-cell imaging for observing subcellular organelles, conventional visible light microscopy remains constrained by the diffraction limit, achieving maximum resolutions between 200 and 250 nanometers [171]. Taking mitochondria as an example, the width of the crest is often 20–100 nm. Therefore, super-resolution microscopy techniques have emerged, including wide-field imaging methods based on spatial spectrum expansion (the representative technology is structured illumination microscopy (SIM)), the laser scanning imaging method based on point spread function (PSF) compression (the representative technology is stimulated emission depletion (STED)), single-molecule localization microscopy (SMLM) based on single-molecule localization imaging method (the representative technology is stochastic optical reconstruction microscopy (STORM)), and photoactivated localization microscopy (PALM) [172]. SIM technology does not require special fluorescent labeling, requires low illumination light intensity, has low photobleaching and photodamage to the sample, and is fast, making it suitable for live-cell imaging, especially long-term live-cell imaging. STED technology relies on the principle of using two lasers, one to excite the fluorescent molecules and the other, the STED laser, to suppress the emission of fluorescent groups located outside the excitation center [173]. As a result, a smaller point spread function is obtained, and its optical resolution is improved. The principle of SMLM technology is to collect sparse fluorescent molecules multiple times. The result of synthetic single-molecule localization is the distribution of fluorescent molecules in the entire field of view, and the position information of these single molecules is superimposed to achieve super-resolution imaging [174]. Combining the advantages of the three techniques, SIM technology is suitable for observing the interaction of mitochondria and other organelles and the division and fusion of mitochondria in a large field of view or with ultra-fast speed; STED technology can specifically label mitochondrial proteins and is suitable for dynamically observing changes in the ultrastructure of mitochondria in a small field of view; SMLM technology is suitable for observing protein localization and interactions. With the vigorous development of artificial intelligence and deep learning, in their latest research, et al. demonstrated a ratiometric HCIO probe AHOH that can simultaneously perform two-color imaging of lysosomes and mitochondria for monitoring mitochondrial autophagy [175]. In addition, a new near-infrared fluorescence probe, HBimmCue, has been developed. Combined with fluorescence lifetime imaging microscopy (FLIM), STED-FLIM imaging is used to reveal changes in mitochondrial membrane order, which in turn reflects mitochondrial function and cellular respiration levels [176,177].
In the realm of single-cell transcriptome sequencing, the single-cell transcriptome represents the comprehensive expression profile of all mRNAs within an individual cell at a specific timepoint, capturing the cell’s functional essence and biological state. Prominent technology packages, such as STRT-seq [178,179], Smart-seq [180,181], CEL-seq [182,183], Quartz-seq [184], and MATQ-seq [185,186], enable precise analysis, while leading platforms like 10x Genomics, BD Rhapsody, and Fluidigm C1 empower researchers with scalable, high-resolution solutions. Single-cell metabolomics is the qualitative and quantitative analysis of small molecule metabolites in biological samples. As direct products of the functional activities of organisms, changes in metabolites can reflect changes in phenotype. It is mainly divided into microscopy-based platforms, spectroscopy-based platforms, and mass spectrometry-based platforms. In comparison, mass spectrometry stands out among these techniques due to its high detection sensitivity, selectivity, wide detection range, fast analysis speed, and strong molecular structure identification capabilities. The process mainly includes single-cell sampling, content measurement, and data analysis. The workflow of single-cell metabolomics data analysis includes data preprocessing, metabolite annotation, statistical analysis, network and pathway analysis, and data visualization [187,188]. However, the single-cell metabolome is highly susceptible to variability in instrument operation and personnel expertise, which can significantly compromise the reliability of results. In terms of single-cell proteomics technology, the total protein content of a single cell is only 100–200 pg, and the amount in an oocyte is even less. Therefore, any loss will have an immeasurable impact, and it also puts forward more stringent requirements for the sensitivity and accuracy of the detection method. Single-cell proteomics can be divided into antibody-based detection methods such as mass spectrometry flow cytometry, PCR-based detection methods such as CITE-seq and REAP-seq, and mass spectrometry-based detection methods such as nanoPOTS. Recently, Yang et al. unveiled groundbreaking evidence identifying NADase CD38 as a central determinant governing age-related deterioration in oocyte quality [171]. At present, mass spectrometry has become the mainstream analytical tool for large-volume proteomics due to its high precision and high-throughput advantages [187]. In this study, researchers employed cutting-edge mass spectrometry-based single-cell proteomics technology, successfully identifying and quantifying a remarkable average of 1200 distinct proteins within each meticulously analyzed mouse oocyte sample [189,190]. In the realm of single-cell epigenomes, methylation predominantly arises at the fifth carbon position of cytosine derivatives, where critical molecular signatures like 5-hydroxymethylcytosine, 5-formylcytosine, 5-carboxycytosine, and m6A are forged through this biochemical artistry. Epigenome sequencing focuses on mining epigenetic information such as genome methylation and epigenetic information such as genome methylation and histone modification regulation. Common techniques encompass revolutionary single-nuclei ATAC-seq and precision-driven CUT&Tag [191]. Researchers constructed genome-wide maps of accessible chromatin in bovine oocytes and early embryos using ATAC-seq, revealing dynamic chromatin accessibility patterns during early embryonic development in cattle. The study demonstrated low chromatin accessibility in bovine oocytes and 2-/4-cell embryos, which markedly increased during the embryonic genome activation (EGA) phase and peaked in day 14 elongated embryos. Robust ATAC-seq signals were detected at both transcription start sites (TSSs) and transcription termination sites (TTSs) [192]. A separate mouse model study revealed that selective chromatin accessibility present in sperm and MII oocytes was extensively erased during early pronuclear stages through a protamine phosphorylation-dependent mechanism [193]. Studies in mouse models employing single-oocyte RNA sequencing with CUT&Tag analysis have demonstrated that DIS3 enzyme mediates transcriptional silencing through intergenic RNA degradation, a critical mechanism regulating chromatin condensation and meiotic resumption [194]. Separate investigations focusing on bovine germ cells have systematically analyzed chromatin accessibility features of activation markers (H3K4me3, H3K27ac) and repression markers (H3K9me3, H3K27me3) across developmental stages, including oocytes; two-cell, four-cell, and eight-cell embryos; morulae; blastocysts; inner cell masses; and trophectoderm tissues [195]. For example, CUT&Tag was used to identify differentially enriched regions of histone H3K23su in 5-resistant HCT15 cells by integrating with ATAC-seq and RNA sequencing data [196]. In summary, the technology for high-resolution observation of subcellular organelles in living cells and single-cell omics sequencing is becoming increasingly mature, which facilitates a better exploration of the link between cellular physiological activities and various intracellular organelles. However, there is no relatively complete report on the link between mitochondrial function and activity, the MQC network and oocyte maturation in bovine oocytes, and the underlying molecular mechanisms remain unclear.

9. Conclusions and Future Perspectives

Mammalian oocyte maturation plays a pivotal role in determining reproductive efficiency and advancing the livestock industry, positioning the enhancement of oocyte quality as a paramount objective in both animal husbandry and life sciences. Cutting-edge research has recently uncovered nutritional compounds and groundbreaking mitochondrial activators—such as resveratrol [197], ubiquinol-10 (the bioactive form of coenzyme Q10), and dehydroepiandrosterone—that demonstrate remarkable efficacy in bolstering mitochondrial efficiency and elevating the developmental potential of oocytes (Table 1) [107]. Coenzyme Q10 supplementation and nicotinamide mononucleotide intake improve ovarian reserve and mitochondrial performance in murine oocytes. Melatonin optimizes mitochondrial calcium homeostasis and membrane potential in human oocytes while upregulating antioxidant gene expression to enhance cytoplasmic maturation in bovine models [197]. The hydroxylamine derivative BGP-15 restores mitochondrial function in oocytes from obese mice, whereas quercetin increases mitochondrial quantity and reduces structural anomalies. Mitoquinone mesylate (MitoQ) effectively lowers ROS levels during bovine oocyte in vitro maturation [38]. Emerging evidence illuminates rhodioloside’s pivotal role in sustaining mitochondrial integrity within aged mouse oocytes, astaxanthin’s remarkable capacity to diminish ROS while revitalizing mitochondrial dynamics in bovine reproductive systems, and cycloastragenol’s groundbreaking telomerase-enhancing prowess that elevates bovine oocyte maturation to unprecedented levels [198,199,200,201].
Despite these advances, the molecular interplay between MQC networks and oocyte maturation remains poorly defined. Current research predominantly employs in vitro maturation models and protein-targeted analyses, leaving critical gaps in understanding how MQC mechanisms—particularly mitochondrial biogenesis, dynamics, and mitophagy—orchestrate maturation processes. Future investigations should prioritize elucidating how MQC-related physiological activities modulate the MQC network, the mechanistic links between MQC networks and bovine oocyte maturation, and the molecular pathways through which mitochondrial biogenesis, dynamics, and mitophagy coordinate to regulate maturation. Resolving these questions will establish targeted therapeutic strategies to optimize oocyte maturation and mitochondrial functionality.

Author Contributions

Conceptualization, D.-J.X. and Y.-R.Z.; writing—original draft preparation, Y.-R.Z.; writing—review and editing, D.-J.X.; project administration and funding acquisition, D.-J.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (32102623), the Fundamental Research Funds for Central Universities (SWU-KT25009).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Liu, S.Y.; Li, Y.C.; Tian, X.Y.; Zhou, Y.; Guo, K.P.; Fan, H.Y.; Liang, X.W.; Ou, X.H.; Sha, Q.Q. A long-acting recombinant FSH supports high-quality mouse follicle development and oocyte maturation in vitro by coordinating somatic and germ cell transcriptomes. Mol. Hum. Reprod. 2023, 29, gaad013. [Google Scholar] [CrossRef] [PubMed]
  2. Dieci, C.; Lodde, V.; Labreque, R.; Dufort, I.; Tessaro, I.; Sirard, M.A.; Luciano, A.M. Differences in cumulus cell gene expression indicate the benefit of a pre-maturation step to improve in-vitro bovine embryo production. Mol. Hum. Reprod. 2016, 22, 882–897. [Google Scholar] [CrossRef] [PubMed]
  3. Zaffagnini, G.; Cheng, S.; Salzer, M.C.; Pernaute, B.; Duran, J.M.; Irimia, M.; Schuh, M.; Böke, E. Mouse oocytes sequester aggregated proteins in degradative super-organelles. Cell 2024, 187, 1109–1126.e1121. [Google Scholar] [CrossRef] [PubMed]
  4. Yan, J.J.; Wang, Y.Y.; Shi, Z.Y.; Ding, Y.Y.; Wen, H.Q.; Wu, M.P.; Sun, S.C.; Cai, Y.F.; Zhang, Y. SIRT5 modulates mitochondria function via mitophagy and antioxidant mechanisms to facilitate oocyte maturation in mice. Int. J. Biol. Macromol. 2025, 306, 141488. [Google Scholar] [CrossRef]
  5. Cai, J.; Li, Y.; Zhao, B.; Bao, Z.; Li, J.; Sun, S.; Chen, Y.; Wu, X. N-Acetylcysteine Alleviates D-Galactose-Induced Injury of Ovarian Granulosa Cells in Female Rabbits by Regulating the PI3K/Akt/mTOR Signaling Pathway. Antioxidants 2024, 13, 384. [Google Scholar] [CrossRef]
  6. Li, C.W.; Ge, W. Spatiotemporal expression of bone morphogenetic protein family ligands and receptors in the zebrafish ovary: A potential paracrine-signaling mechanism for oocyte-follicle cell communication. Biol. Reprod. 2011, 85, 977–986. [Google Scholar] [CrossRef]
  7. Marei, W.F.A.; Van den Bosch, L.; Pintelon, I.; Mohey-Elsaeed, O.; Bols, P.E.J.; Leroy, J. Mitochondria-targeted therapy rescues development and quality of embryos derived from oocytes matured under oxidative stress conditions: A bovine in vitro model. Hum. Reprod. 2019, 34, 1984–1998. [Google Scholar] [CrossRef]
  8. Kumar, K.; Venturas, M.; Needleman, D.J.; Racowsky, C.; Wells, D. Extensive analysis of mitochondrial DNA quantity and sequence variation in human cumulus cells and assisted reproduction outcomes. Hum. Reprod. 2021, 37, 66–79. [Google Scholar] [CrossRef]
  9. Yang, Q.; Xi, Q.; Wang, M.; Long, R.; Hu, J.; Li, Z.; Ren, X.; Zhu, L.; Jin, L. Rapamycin improves the quality and developmental competence of mice oocytes by promoting DNA damage repair during in vitro maturation. Reprod. Biol. Endocrinol. 2022, 20, 67. [Google Scholar] [CrossRef]
  10. Bilodeau-Goeseels, S.; Panich, P.L.; Kastelic, J.P. Activation of AMP-activated protein kinase may not be involved in AICAR- and metformin-mediated meiotic arrest in bovine denuded and cumulus-enclosed oocytes in vitro. Zygote 2011, 19, 97–106. [Google Scholar] [CrossRef]
  11. Santiquet, N.; Sasseville, M.; Laforest, M.; Guillemette, C.; Gilchrist, R.B.; Richard, F.J. Activation of 5’adenosine monophosphate-activated protein kinase blocks cumulus cell expansion through inhibition of protein synthesis during in vitro maturation in Swine. Biol. Reprod. 2014, 91, 51. [Google Scholar] [CrossRef] [PubMed]
  12. Ran, Z.; Liu, R.; Shi, H.; Wang, X.; Wu, Z.; Zhou, S.; Liao, J.; Hu, L.; Hu, Y.; Zhou, J.; et al. mTOR signaling mediates energy metabolic equilibrium in bovine and mouse oocytes during the ovulatory phase†. Biol. Reprod. 2025, 112, 474–484. [Google Scholar] [CrossRef] [PubMed]
  13. Shi, L.; Ying, H.; Dai, Y.; Rong, Y.; Chen, J.; Zhou, F.; Wang, S.; Xu, S.; Tong, X.; Zhang, S. Upregulated let-7 expression in the follicular fluid of patients with endometriomas leads to dysfunction of granulosa cells through targeting of IGF1R. Hum. Reprod. 2025, 40, 119–137. [Google Scholar] [CrossRef] [PubMed]
  14. Cunningham, J.T.; Rodgers, J.T.; Arlow, D.H.; Vazquez, F.; Mootha, V.K.; Puigserver, P. mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex. Nature 2007, 450, 736–740. [Google Scholar] [CrossRef]
  15. Ramanathan, A.; Schreiber, S.L. Direct control of mitochondrial function by mTOR. Proc. Natl. Acad. Sci. USA 2009, 106, 22229–22232. [Google Scholar] [CrossRef]
  16. Morita, M.; Prudent, J.; Basu, K.; Goyon, V.; Katsumura, S.; Hulea, L.; Pearl, D.; Siddiqui, N.; Strack, S.; McGuirk, S.; et al. mTOR Controls Mitochondrial Dynamics and Cell Survival via MTFP1. Mol. Cell 2017, 67, 922–935.e925. [Google Scholar] [CrossRef]
  17. El-Sheikh, M.; Mesalam, A.A.; Kang, S.M.; Joo, M.D.; Soliman, S.S.; Khalil, A.A.K.; Ahn, M.J.; Kong, I.K. Modulation of Apoptosis and Autophagy by Melatonin in Juglone-Exposed Bovine Oocytes. Animals 2023, 13, 1475. [Google Scholar] [CrossRef]
  18. El-Sheikh, M.; Mesalam, A.; Khalil, A.A.K.; Idrees, M.; Ahn, M.J.; Mesalam, A.A.; Kong, I.K. Downregulation of PI3K/AKT/mTOR Pathway in Juglone-Treated Bovine Oocytes. Antioxidants 2023, 12, 114. [Google Scholar] [CrossRef]
  19. Ellederová, Z.; Cais, O.; Susor, A.; Uhlírová, K.; Kovárová, H.; Jelínková, L.; Tomek, W.; Kubelka, M. ERK1/2 map kinase metabolic pathway is responsible for phosphorylation of translation initiation factor eIF4E during in vitro maturation of pig oocytes. Mol. Reprod. Dev. 2008, 75, 309–317. [Google Scholar] [CrossRef]
  20. Tomek, W.; Melo Sterza, F.A.; Kubelka, M.; Wollenhaupt, K.; Torner, H.; Anger, M.; Kanitz, W. Regulation of translation during in vitro maturation of bovine oocytes: The role of MAP kinase, eIF4E (cap binding protein) phosphorylation, and eIF4E-BP1. Biol. Reprod. 2002, 66, 1274–1282. [Google Scholar] [CrossRef]
  21. Jansova, D.; Koncicka, M.; Tetkova, A.; Cerna, R.; Malik, R.; Del Llano, E.; Kubelka, M.; Susor, A. Regulation of 4E-BP1 activity in the mammalian oocyte. Cell Cycle 2017, 16, 927–939. [Google Scholar] [CrossRef] [PubMed]
  22. Mayer, S.; Wrenzycki, C.; Tomek, W. Inactivation of mTor arrests bovine oocytes in the metaphase-I stage, despite reversible inhibition of 4E-BP1 phosphorylation. Mol. Reprod. Dev. 2014, 81, 363–375. [Google Scholar] [CrossRef] [PubMed]
  23. Cotterill, M.; Harris, S.E.; Collado Fernandez, E.; Lu, J.; Huntriss, J.D.; Campbell, B.K.; Picton, H.M. The activity and copy number of mitochondrial DNA in ovine oocytes throughout oogenesis in vivo and during oocyte maturation in vitro. Mol. Hum. Reprod. 2013, 19, 444–450. [Google Scholar] [CrossRef] [PubMed]
  24. Ge, H.; Tollner, T.L.; Hu, Z.; Dai, M.; Li, X.; Guan, H.; Shan, D.; Zhang, X.; Lv, J.; Huang, C.; et al. The importance of mitochondrial metabolic activity and mitochondrial DNA replication during oocyte maturation in vitro on oocyte quality and subsequent embryo developmental competence. Mol. Reprod. Dev. 2012, 79, 392–401. [Google Scholar] [CrossRef]
  25. Jeseta, M.; Ctvrtlikova Knitlova, D.; Hanzalova, K.; Hulinska, P.; Hanulakova, S.; Milakovic, I.; Nemcova, L.; Kanka, J.; Machatkova, M. Mitochondrial patterns in bovine oocytes with different meiotic competence related to their in vitro maturation. Reprod. Domest. Anim. 2014, 49, 469–475. [Google Scholar] [CrossRef]
  26. Němcová, L.; Hulínská, P.; Ješeta, M.; Kempisty, B.; Kaňka, J.; Machatková, M. Expression of selected mitochondrial genes during in vitro maturation of bovine oocytes related to their meiotic competence. Theriogenology 2019, 133, 104–112. [Google Scholar] [CrossRef]
  27. Wang, X.H.; Yin, S.; Ou, X.H.; Luo, S.M. Increase of mitochondria surrounding spindle causes mouse oocytes arrested at metaphase I stage. Biochem. Biophys. Res. Commun. 2020, 527, 1043–1049. [Google Scholar] [CrossRef]
  28. Pan, Z.N.; Zhang, H.L.; Zhang, K.H.; Ju, J.Q.; Liu, J.C.; Sun, S.C. Insufficient MIRO1 contributes to declined oocyte quality during reproductive aging. Sci. China-Life Sci. 2025, 68, 764–776. [Google Scholar] [CrossRef]
  29. Di Rienzo, M.; Romagnoli, A.; Refolo, G.; Vescovo, T.; Ciccosanti, F.; Zuchegna, C.; Lozzi, F.; Occhigrossi, L.; Piacentini, M.; Fimia, G.M. Role of AMBRA1 in mitophagy regulation: Emerging evidence in aging-related diseases. Autophagy 2024, 20, 2602–2615. [Google Scholar] [CrossRef]
  30. Liu, Y.P.; He, B.; Wang, W.X.; Pan, W.L.; Jiao, L.; Yan, J.J.; Sun, S.C.; Zhang, Y. PKD regulates mitophagy to prevent oxidative stress and mitochondrial dysfunction during mouse oocyte maturation. Mitochondrion 2024, 78, 101946. [Google Scholar] [CrossRef]
  31. Dudkina, N.V.; Eubel, H.; Keegstra, W.; Boekema, E.J.; Braun, H.P. Structure of a mitochondrial supercomplex formed by respiratory-chain complexes I and III. Proc. Natl. Acad. Sci. USA 2005, 102, 3225–3229. [Google Scholar] [CrossRef]
  32. Magalhaes-Novais, S.; Blecha, J.; Naraine, R.; Mikesova, J.; Abaffy, P.; Pecinova, A.; Milosevic, M.; Bohuslavova, R.; Prochazka, J.; Khan, S.; et al. Mitochondrial respiration supports autophagy to provide stress resistance during quiescence. Autophagy 2022, 18, 2409–2426. [Google Scholar] [CrossRef] [PubMed]
  33. Khan, A.U.H.; Rathore, M.G.; Allende-Vega, N.; Vo, D.N.; Belkhala, S.; Orecchioni, S.; Talarico, G.; Bertolini, F.; Cartron, G.; Lecellier, C.H.; et al. Human Leukemic Cells performing Oxidative Phosphorylation (OXPHOS) Generate an Antioxidant Response Independently of Reactive Oxygen species (ROS) Production. EBioMedicine 2016, 3, 43–53. [Google Scholar] [CrossRef] [PubMed]
  34. Coticchio, G.; Dal Canto, M.; Mignini Renzini, M.; Guglielmo, M.C.; Brambillasca, F.; Turchi, D.; Novara, P.V.; Fadini, R. Oocyte maturation: Gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum. Reprod. Update 2015, 21, 427–454. [Google Scholar] [CrossRef]
  35. Richani, D.; Gilchrist, R.B. The epidermal growth factor network: Role in oocyte growth, maturation and developmental competence. Hum. Reprod. Update 2018, 24, 1–14. [Google Scholar] [CrossRef]
  36. Gu, L.; Wang, Q.; Sun, Q.Y. Histone modifications during mammalian oocyte maturation: Dynamics, regulation and functions. Cell Cycle 2010, 9, 1942–1950. [Google Scholar] [CrossRef]
  37. Adhikari, D.; Lee, I.W.; Yuen, W.S.; Carroll, J. Oocyte mitochondria-key regulators of oocyte function and potential therapeutic targets for improving fertility. Biol. Reprod. 2022, 106, 366–377. [Google Scholar] [CrossRef]
  38. Kirillova, A.; Smitz, J.E.J.; Sukhikh, G.T.; Mazunin, I. The Role of Mitochondria in Oocyte Maturation. Cells 2021, 10, 2484. [Google Scholar] [CrossRef]
  39. Giotto, A.B.; Brum, D.D.; Santos, F.W.; Guimaraes, A.C.G.; Gonçalves, C.G.M.; Pavin, C.U.M.; Folchini, N.P.; Moyses, A.B.; Missio, D.; Leivas, F.G. Oxygen tension and oocyte density during in vitro maturation affect the in vitro fertilization of bovine oocytes. Semin.-Cienc. Agrar. 2015, 36, 4277–4287. [Google Scholar] [CrossRef]
  40. Yang, X.; Liu, Q.; Li, Y.; Tang, Q.; Wu, T.; Chen, L.; Pu, S.; Zhao, Y.; Zhang, G.; Huang, C.; et al. The diabetes medication canagliflozin promotes mitochondrial remodelling of adipocyte via the AMPK-Sirt1-Pgc-1α signalling pathway. Adipocyte 2020, 9, 484–494. [Google Scholar] [CrossRef]
  41. Scarpulla, R.C. Nuclear control of respiratory gene expression in mammalian cells. J. Cell. Biochem. 2006, 97, 673–683. [Google Scholar] [CrossRef] [PubMed]
  42. Quirós, P.M.; Langer, T.; López-Otín, C. New roles for mitochondrial proteases in health, ageing and disease. Nat. Rev. Mol. Cell Biol. 2015, 16, 345–359. [Google Scholar] [CrossRef] [PubMed]
  43. Archer, S.L. Mitochondrial dynamics--mitochondrial fission and fusion in human diseases. N. Engl. J. Med. 2013, 369, 2236–2251. [Google Scholar] [CrossRef] [PubMed]
  44. Strauss, K.A.; Jinks, R.N.; Puffenberger, E.G.; Venkatesh, S.; Singh, K.; Cheng, I.; Mikita, N.; Thilagavathi, J.; Lee, J.; Sarafianos, S.; et al. CODAS syndrome is associated with mutations of LONP1, encoding mitochondrial AAA+ Lon protease. Am. J. Hum. Genet. 2015, 96, 121–135. [Google Scholar] [CrossRef]
  45. Bao, F.; Zhou, L.; Zhou, R.; Huang, Q.; Chen, J.; Zeng, S.; Wu, Y.; Yang, L.; Qian, S.; Wang, M.; et al. Mitolysosome exocytosis, a mitophagy-independent mitochondrial quality control in flunarizine-induced parkinsonism-like symptoms. Sci. Adv. 2022, 8, eabk2376. [Google Scholar] [CrossRef]
  46. Lu, Y.; Li, Z.; Zhang, S.; Zhang, T.; Liu, Y.; Zhang, L. Cellular mitophagy: Mechanism, roles in diseases and small molecule pharmacological regulation. Theranostics 2023, 13, 736–766. [Google Scholar] [CrossRef]
  47. Qi, X.Y.; Yuan, J.D.; Liu, Z.Y.; Jiang, X.Q.; Zhang, Q.; Zhang, S.L.; Zhao, L.; Ke, L.Y.; Zhang, C.Y.; Li, Y.; et al. Sirtuin 3-mediated deacetylation of superoxide dismutase 2 ameliorates sodium fluoride-induced mitochondrial dysfunction in porcine oocytes. Sci. Total Environ. 2024, 908, 168306. [Google Scholar] [CrossRef]
  48. Shen, Y.; Wu, Q.; Shi, J.; Zhou, S. Regulation of SIRT3 on mitochondrial functions and oxidative stress in Parkinson’s disease. Biomed. Pharmacother. 2020, 132, 110928. [Google Scholar] [CrossRef]
  49. Li, X.; Duan, J.; Wang, S.; Cheng, J.; Chen, H.; Zhang, Z.; Yang, L.; Hua, R.; Li, Q. Isorhamnetin protects porcine oocytes from zearalenone-induced reproductive toxicity through the PI3K/Akt signaling pathway. J. Anim. Sci. Biotechnol. 2023, 14, 22. [Google Scholar] [CrossRef]
  50. Alcantara da Silva, J.V.; Ispada, J.; Nociti, R.P.; da Fonseca Junior, A.M.; de Lima, C.B.; Dos Santos, E.C.; Chiaratti, M.R.; Milazzotto, M.P. The central role of pyruvate metabolism on the epigenetic maturation and transcriptional profile of bovine oocytes. Reproduction 2024, 167, e230181. [Google Scholar] [CrossRef]
  51. Richani, D.; Dunning, K.R.; Thompson, J.G.; Gilchrist, R.B. Metabolic co-dependence of the oocyte and cumulus cells: Essential role in determining oocyte developmental competence. Hum. Reprod. Update 2021, 27, 27–47. [Google Scholar] [CrossRef] [PubMed]
  52. Dunning, K.R.; Robker, R.L. Promoting lipid utilization with l-carnitine to improve oocyte quality. Anim. Reprod. Sci. 2012, 134, 69–75. [Google Scholar] [CrossRef] [PubMed]
  53. Dunning, K.R.; Akison, L.K.; Russell, D.L.; Norman, R.J.; Robker, R.L. Increased beta-oxidation and improved oocyte developmental competence in response to l-carnitine during ovarian in vitro follicle development in mice. Biol. Reprod. 2011, 85, 548–555. [Google Scholar] [CrossRef]
  54. Leitão, A.M.F.; Silva, B.R.; Barbalho, E.C.; Paulino, L.R.M.; Costa, F.D.C.; Martins, F.S.; Silva, J.R.V. The role of L-carnitine in the control of oxidative stress and lipid β-oxidation during in vitro follicle growth, oocyte maturation, embryonic development and cryopreservation: A review. Zygote 2024, 32, 335–340. [Google Scholar] [CrossRef]
  55. Xu, D.; Li, C.; Huang, Y.; Hu, K.; Wang, C.; Zhou, P.; Shen, H.; Liu, C.; Xu, J.; He, J.; et al. Ferric ammonium citrate regulates iron death in mature porcine oocytes and their embryonic development in vitro through the NRF2 signaling pathway. Theriogenology 2025, 232, 1–8. [Google Scholar] [CrossRef]
  56. Colpman, P.; Dasgupta, A.; Archer, S.L. The Role of Mitochondrial Dynamics and Mitotic Fission in Regulating the Cell Cycle in Cancer and Pulmonary Arterial Hypertension: Implications for Dynamin-Related Protein 1 and Mitofusin2 in Hyperproliferative Diseases. Cells 2023, 12, 1897. [Google Scholar] [CrossRef]
  57. Ago, T.; Yeh, I.; Yamamoto, M.; Schinke-Braun, M.; Brown, J.A.; Tian, B.; Sadoshima, J. Thioredoxin1 upregulates mitochondrial proteins related to oxidative phosphorylation and TCA cycle in the heart. Antioxid. Redox Signal. 2006, 8, 1635–1650. [Google Scholar] [CrossRef]
  58. Li, J.; Yang, D.; Li, Z.; Zhao, M.; Wang, D.; Sun, Z.; Wen, P.; Dai, Y.; Gou, F.; Ji, Y.; et al. PINK1/Parkin-mediated mitophagy in neurodegenerative diseases. Ageing Res. Rev. 2023, 84, 101817. [Google Scholar] [CrossRef]
  59. Nguyen, T.N.; Padman, B.S.; Lazarou, M. Deciphering the Molecular Signals of PINK1/Parkin Mitophagy. Trends Cell Biol. 2016, 26, 733–744. [Google Scholar] [CrossRef]
  60. Tang, J.; Peng, W.; Ji, J.; Peng, C.; Wang, T.; Yang, P.; Gu, J.; Feng, Y.; Jin, K.; Wang, X.; et al. GPR176 Promotes Cancer Progression by Interacting with G Protein GNAS to Restrain Cell Mitophagy in Colorectal Cancer. Adv. Sci. 2023, 10, e2205627. [Google Scholar] [CrossRef]
  61. Chen, X.; Ji, Y.; Liu, R.; Zhu, X.; Wang, K.; Yang, X.; Liu, B.; Gao, Z.; Huang, Y.; Shen, Y.; et al. Mitochondrial dysfunction: Roles in skeletal muscle atrophy. J. Transl. Med. 2023, 21, 503. [Google Scholar] [CrossRef] [PubMed]
  62. Xue, Y.; Cheng, X.; Ma, Z.Q.; Wang, H.P.; Zhou, C.; Li, J.; Zhang, D.L.; Hu, L.L.; Cui, Y.F.; Huang, J.; et al. Polystyrene nanoplastics induce apoptosis, autophagy, and steroidogenesis disruption in granulosa cells to reduce oocyte quality and fertility by inhibiting the PI3K/AKT pathway in female mice. J. Nanobiotechnol. 2024, 22, 460. [Google Scholar] [CrossRef] [PubMed]
  63. Zhou, C.J.; Wang, X.Y.; Dong, Y.H.; Wang, D.H.; Han, Z.; Zhang, X.J.; Sun, Q.Y.; Carroll, J.; Liang, C.G. CENP-F-dependent DRP1 function regulates APC/C activity during oocyte meiosis I. Nat. Commun. 2022, 13, 7732. [Google Scholar] [CrossRef] [PubMed]
  64. Scarpulla, R.C. Transcriptional paradigms in mammalian mitochondrial biogenesis and function. Physiol. Rev. 2008, 88, 611–638. [Google Scholar] [CrossRef]
  65. Fan, S.; Zhao, X.; Xie, W.; Yang, X.; Yu, W.; Tang, Z.; Chen, Y.; Yuan, Z.; Han, Y.; Sheng, X.; et al. The effect of 3-Methyl-4-Nitrophenol on the early ovarian follicle development in mice by disrupting the clock genes expression. Chem. Biol. Interact. 2022, 363, 110001. [Google Scholar] [CrossRef]
  66. Ma, C.; Xu, Y.; Zhang, X.; Shi, X.; Zhang, Y.; Luo, M.; Wu, C.; Ding, Z.; Xiang, H.; Cao, Y. Melatonin mitigates PNMC-induced disruption of spindle assembly and mitochondrial function in mouse Oocytes. Ecotoxicol. Environ. Saf. 2024, 282, 116703. [Google Scholar] [CrossRef]
  67. Zhang, Z.; Jia, Z. Pre-IVM with C-type natriuretic peptide promotes mitochondrial biogenesis of bovine oocytes via activation of CREB. Sci. Rep. 2024, 14, 16260. [Google Scholar] [CrossRef]
  68. Jia, Z.; Yang, X.; Liu, K. Treatment of cattle oocytes with C-type natriuretic peptide before in vitro maturation enhances oocyte mitochondrial function. Anim. Reprod. Sci. 2021, 225, 106685. [Google Scholar] [CrossRef]
  69. Ardehjani, N.A.; Agha-Hosseini, M.; Nashtaei, M.S.; Khodarahmian, M.; Shabani, M.; Jabarpour, M.; Fereidouni, F.; Rastegar, T.; Amidi, F. Resveratrol ameliorates mitochondrial biogenesis and reproductive outcomes in women with polycystic ovary syndrome undergoing assisted reproduction: A randomized, triple-blind, placebo-controlled clinical trial. J. Ovarian Res. 2024, 17, 143. [Google Scholar] [CrossRef]
  70. Gherardi, G.; Corbioli, G.; Ruzza, F.; Rizzuto, R. CoQ(10) and Resveratrol Effects to Ameliorate Aged-Related Mitochondrial Dysfunctions. Nutrients 2022, 14, 4326. [Google Scholar] [CrossRef]
  71. Itami, N.; Shirasuna, K.; Kuwayama, T.; Iwata, H. Short-term heat stress induces mitochondrial degradation and biogenesis and enhances mitochondrial quality in porcine oocytes. J. Therm. Biol. 2018, 74, 256–263. [Google Scholar] [CrossRef] [PubMed]
  72. Roth, Z. Symposium review: Reduction in oocyte developmental competence by stress is associated with alterations in mitochondrial function. J. Dairy Sci. 2018, 101, 3642–3654. [Google Scholar] [CrossRef] [PubMed]
  73. Niu, Y.J.; Zhou, W.; Nie, Z.W.; Shin, K.T.; Cui, X.S. Melatonin enhances mitochondrial biogenesis and protects against rotenone-induced mitochondrial deficiency in early porcine embryos. J. Pineal Res. 2020, 68, e12627. [Google Scholar] [CrossRef] [PubMed]
  74. Sánchez, F.; Romero, S.; De Vos, M.; Verheyen, G.; Smitz, J. Human cumulus-enclosed germinal vesicle oocytes from early antral follicles reveal heterogeneous cellular and molecular features associated with in vitro maturation capacity. Hum. Reprod. 2015, 30, 1396–1409. [Google Scholar] [CrossRef]
  75. Xu, D.; Wu, L.; Jiang, X.; Yang, L.; Cheng, J.; Chen, H.; Hua, R.; Geng, G.; Yang, L.; Li, Q. SIRT2 Inhibition Results in Meiotic Arrest, Mitochondrial Dysfunction, and Disturbance of Redox Homeostasis during Bovine Oocyte Maturation. Int. J. Mol. Sci. 2019, 20, 1365. [Google Scholar] [CrossRef]
  76. Zhang, L.; Hou, X.; Ma, R.; Moley, K.; Schedl, T.; Wang, Q. Sirt2 functions in spindle organization and chromosome alignment in mouse oocyte meiosis. FASEB J. 2014, 28, 1435–1445. [Google Scholar] [CrossRef]
  77. Ferreira, A.F.; Soares, M.; Almeida-Santos, T.; Ramalho-Santos, J.; Sousa, A.P. Aging and oocyte competence: A molecular cell perspective. WIREs Mech. Dis. 2023, 15, e1613. [Google Scholar] [CrossRef]
  78. Mikwar, M.; MacFarlane, A.J.; Marchetti, F. Mechanisms of oocyte aneuploidy associated with advanced maternal age. Mutat. Res. Rev. Mutat. Res. 2020, 785, 108320. [Google Scholar] [CrossRef]
  79. Cantó, C.; Gerhart-Hines, Z.; Feige, J.N.; Lagouge, M.; Noriega, L.; Milne, J.C.; Elliott, P.J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 2009, 458, 1056–1060. [Google Scholar] [CrossRef]
  80. Gómora-García, J.C.; Montiel, T.; Hüttenrauch, M.; Salcido-Gómez, A.; García-Velázquez, L.; Ramiro-Cortés, Y.; Gomora, J.C.; Castro-Obregón, S.; Massieu, L. Effect of the Ketone Body, D-β-Hydroxybutyrate, on Sirtuin2-Mediated Regulation of Mitochondrial Quality Control and the Autophagy-Lysosomal Pathway. Cells 2023, 12, 486. [Google Scholar] [CrossRef]
  81. Cheng, Z. FoxO transcription factors in mitochondrial homeostasis. Biochem. J. 2022, 479, 525–536. [Google Scholar] [CrossRef]
  82. Chao, X.; Wang, S.; Zhao, K.; Li, Y.; Williams, J.A.; Li, T.; Chavan, H.; Krishnamurthy, P.; He, X.C.; Li, L.; et al. Impaired TFEB-Mediated Lysosome Biogenesis and Autophagy Promote Chronic Ethanol-Induced Liver Injury and Steatosis in Mice. Gastroenterology 2018, 155, 865–879.e812. [Google Scholar] [CrossRef]
  83. Perales, J.A.; Lawan, A.; Bajpeyi, S.; Han, S.M.; Bennett, A.M.; Min, K. MAP Kinase Phosphatase-5 Deficiency Improves Endurance Exercise Capacity. Cells 2025, 14, 410. [Google Scholar] [CrossRef] [PubMed]
  84. Liu, S.; Wang, Y.; Yang, H.; Tan, J.; Zhang, J.; Zi, D. Pyrroloquinoline quinone promotes human mesenchymal stem cell-derived mitochondria to improve premature ovarian insufficiency in mice through the SIRT1/ATM/p53 pathway. Stem Cell Res. Ther. 2024, 15, 97. [Google Scholar] [CrossRef]
  85. Hao, X.; Zhao, J.; Rodriguez-Wallberg, K.A. Comprehensive atlas of mitochondrial distribution and dynamics during oocyte maturation in mouse models. Biomark. Res. 2024, 12, 125. [Google Scholar] [CrossRef] [PubMed]
  86. Lan, Y.; Zhang, S.; Gong, F.; Lu, C.; Lin, G.; Hu, L. The mitochondrial DNA copy number of cumulus granulosa cells may be related to the maturity of oocyte cytoplasm. Hum. Reprod. 2020, 35, 1120–1129. [Google Scholar] [CrossRef]
  87. Sandoval, H.; Yao, C.K.; Chen, K.; Jaiswal, M.; Donti, T.; Lin, Y.Q.; Bayat, V.; Xiong, B.; Zhang, K.; David, G.; et al. Mitochondrial fusion but not fission regulates larval growth and synaptic development through steroid hormone production. Elife 2014, 3, e03558. [Google Scholar] [CrossRef]
  88. Piao, L.; Fang, Y.H.; Fisher, M.; Hamanaka, R.B.; Ousta, A.; Wu, R.X.; Mutlu, G.M.; Garcia, A.J.; Archer, S.L.; Sharp, W.W. Dynamin-related protein 1 is a critical regulator of mitochondrial calcium homeostasis during myocardial ischemia/reperfusion injury. FASEB J. 2024, 38, e23379. [Google Scholar] [CrossRef]
  89. Zhao, L.; Lu, T.; Gao, L.; Fu, X.; Zhu, S.; Hou, Y. Enriched endoplasmic reticulum-mitochondria interactions result in mitochondrial dysfunction and apoptosis in oocytes from obese mice. J. Anim. Sci. Biotechnol. 2017, 8, 62. [Google Scholar] [CrossRef]
  90. Wu, S.; Lu, Q.; Ding, Y.; Wu, Y.; Qiu, Y.; Wang, P.; Mao, X.; Huang, K.; Xie, Z.; Zou, M.H. Hyperglycemia-Driven Inhibition of AMP-Activated Protein Kinase α2 Induces Diabetic Cardiomyopathy by Promoting Mitochondria-Associated Endoplasmic Reticulum Membranes In Vivo. Circulation 2019, 139, 1913–1936. [Google Scholar] [CrossRef]
  91. Wang, F.; Meng, T.G.; Li, J.; Hou, Y.; Luo, S.M.; Schatten, H.; Sun, Q.Y.; Ou, X.H. Mitochondrial Ca(2 +) Is Related to Mitochondrial Activity and Dynamic Events in Mouse Oocytes. Front. Cell Dev. Biol. 2020, 8, 585932. [Google Scholar] [CrossRef]
  92. Boucret, L.; Chao de la Barca, J.M.; Morinière, C.; Desquiret, V.; Ferré-L’Hôtellier, V.; Descamps, P.; Marcaillou, C.; Reynier, P.; Procaccio, V.; May-Panloup, P. Relationship between diminished ovarian reserve and mitochondrial biogenesis in cumulus cells. Hum. Reprod. 2015, 30, 1653–1664. [Google Scholar] [CrossRef] [PubMed]
  93. Machiela, E.; Liontis, T.; Dues, D.J.; Rudich, P.D.; Traa, A.; Wyman, L.; Kaufman, C.; Cooper, J.F.; Lew, L.; Nadarajan, S.; et al. Disruption of mitochondrial dynamics increases stress resistance through activation of multiple stress response pathways. FASEB J. 2020, 34, 8475–8492. [Google Scholar] [CrossRef] [PubMed]
  94. Zhu, J.; Yang, Q.; Li, H.; Wang, Y.; Jiang, Y.; Wang, H.; Cong, L.; Xu, J.; Shen, Z.; Chen, W.; et al. Sirt3 deficiency accelerates ovarian senescence without affecting spermatogenesis in aging mice. Free Radic. Biol. Med. 2022, 193, 511–525. [Google Scholar] [CrossRef]
  95. Han, T.; Zhao, Y.; Jiao, A.; Sun, Z.; Zhang, H.; Zhao, D.; Wang, H.; Gao, Q. OPA1 deficiency induces mitophagy through PINK1/Parkin pathway during bovine oocytes maturation. Theriogenology 2025, 234, 51–63. [Google Scholar] [CrossRef]
  96. Martinez, A.; Sanchez-Martinez, A.; Pickering, J.T.; Twyning, M.J.; Terriente-Felix, A.; Chen, P.L.; Chen, C.H.; Whitworth, A.J. Mitochondrial CISD1/Cisd accumulation blocks mitophagy and genetic or pharmacological inhibition rescues neurodegenerative phenotypes in Pink1/parkin models. Mol. Neurodegener. 2024, 19, 12. [Google Scholar] [CrossRef]
  97. Gao, Y.; Dong, R.; Yan, J.; Chen, H.; Sang, L.; Yao, X.; Fan, D.; Wang, X.; Zuo, X.; Zhang, X.; et al. Mitochondrial deoxyguanosine kinase is required for female fertility in mice. Acta Biochim. Biophys. Sin. 2024, 56, 427–439. [Google Scholar] [CrossRef]
  98. Van Der Kelen, A.; Li Piani, L.; Mertens, J.; Regin, M.; Couvreu de Deckersberg, E.; Van de Velde, H.; Sermon, K.; Tournaye, H.; Verpoest, W.; Hes, F.J.; et al. The interplay between mitochondrial DNA genotypes, female infertility, ovarian response, and mutagenesis in oocytes. Hum. Reprod. Open 2025, 2025, hoae074. [Google Scholar] [CrossRef]
  99. Liu, X.M.; Zhang, Y.L.; Ji, S.Y.; Zhao, L.W.; Shang, W.N.; Li, D.; Chen, Z.; Tong, C.; Fan, H.Y. Mitochondrial Function Regulated by Mitoguardin-1/2 Is Crucial for Ovarian Endocrine Functions and Ovulation. Endocrinology 2017, 158, 3988–3999. [Google Scholar] [CrossRef]
  100. Zhang, Y.; Liu, X.; Bai, J.; Tian, X.; Zhao, X.; Liu, W.; Duan, X.; Shang, W.; Fan, H.Y.; Tong, C. Mitoguardin Regulates Mitochondrial Fusion through MitoPLD and Is Required for Neuronal Homeostasis. Mol. Cell 2016, 61, 111–124. [Google Scholar] [CrossRef]
  101. Lee, S.H.; Sun, M.H.; Zhou, D.; Jiang, W.J.; Li, X.H.; Heo, G.; Cui, X.S. High Temperature Disrupts Organelle Distribution and Functions Affecting Meiotic Maturation in Porcine Oocytes. Front. Cell Dev. Biol. 2022, 10, 826801. [Google Scholar] [CrossRef] [PubMed]
  102. Park, J.H.; Burgess, J.D.; Faroqi, A.H.; DeMeo, N.N.; Fiesel, F.C.; Springer, W.; Delenclos, M.; McLean, P.J. Alpha-synuclein-induced mitochondrial dysfunction is mediated via a sirtuin 3-dependent pathway. Mol. Neurodegener. 2020, 15, 5. [Google Scholar] [CrossRef]
  103. Garcia, B.M.; Machado, T.S.; Carvalho, K.F.; Nolasco, P.; Nociti, R.P.; Del Collado, M.; Capo Bianco, M.J.D.; Grejo, M.P.; Augusto Neto, J.D.; Sugiyama, F.H.C.; et al. Mice born to females with oocyte-specific deletion of mitofusin 2 have increased weight gain and impaired glucose homeostasis. Mol. Hum. Reprod. 2020, 26, 938–952. [Google Scholar] [CrossRef] [PubMed]
  104. Naón, D.; Hernández-Alvarez, M.I.; Shinjo, S.; Wieczor, M.; Ivanova, S.; Martins de Brito, O.; Quintana, A.; Hidalgo, J.; Palacín, M.; Aparicio, P.; et al. Splice variants of mitofusin 2 shape the endoplasmic reticulum and tether it to mitochondria. Science 2023, 380, eadh9351. [Google Scholar] [CrossRef]
  105. Zhou, G.; Liu, A.; Bai, J.; Liu, H.; Zhu, Y.; Luo, Y.; Zheng, L.; Hou, Y.; Li, J.; Fu, X. Decreased ATF5 level contributes to improved mitochondrial function in oocytes exposed to vitrification stress. Front. Cell Dev. Biol. 2024, 12, 1431683. [Google Scholar] [CrossRef]
  106. Zhang, D.; Fang, X.; Xia, W.; Sun, Q.; Zhang, X.; Qi, Y.; Yu, Y.; Zhou, Z.; Du, D.; Tao, C.; et al. Rutin enhances mitochondrial function and improves the developmental potential of vitrified ovine GV-stage oocyte. Theriogenology 2024, 229, 214–224. [Google Scholar] [CrossRef]
  107. Shen, Q.; Liu, Y.; Li, H.; Zhang, L. Effect of mitophagy in oocytes and granulosa cells on oocyte quality†. Biol. Reprod. 2021, 104, 294–304. [Google Scholar] [CrossRef]
  108. May-Panloup, P.; Boucret, L.; Chao de la Barca, J.M.; Desquiret-Dumas, V.; Ferré-L’Hotellier, V.; Morinière, C.; Descamps, P.; Procaccio, V.; Reynier, P. Ovarian ageing: The role of mitochondria in oocytes and follicles. Hum. Reprod. Update 2016, 22, 725–743. [Google Scholar] [CrossRef]
  109. Li, A.; Gao, M.; Liu, B.; Qin, Y.; Chen, L.; Liu, H.; Wu, H.; Gong, G. Mitochondrial autophagy: Molecular mechanisms and implications for cardiovascular disease. Cell Death Dis. 2022, 13, 444. [Google Scholar] [CrossRef]
  110. Uoselis, L.; Nguyen, T.N.; Lazarou, M. Mitochondrial degradation: Mitophagy and beyond. Mol. Cell 2023, 83, 3404–3420. [Google Scholar] [CrossRef]
  111. Imberechts, D.; Kinnart, I.; Wauters, F.; Terbeek, J.; Manders, L.; Wierda, K.; Eggermont, K.; Madeiro, R.F.; Sue, C.; Verfaillie, C.; et al. DJ-1 is an essential downstream mediator in PINK1/parkin-dependent mitophagy. Brain 2022, 145, 4368–4384. [Google Scholar] [CrossRef] [PubMed]
  112. Ouni, E.; Bouzin, C.; Dolmans, M.M.; Marbaix, E.; Pyr Dit Ruys, S.; Vertommen, D.; Amorim, C.A. Spatiotemporal changes in mechanical matrisome components of the human ovary from prepuberty to menopause. Hum. Reprod. 2020, 35, 1391–1410. [Google Scholar] [CrossRef] [PubMed]
  113. Xu, J.; Sun, L.; Wu, C.; Zhang, S.; Ju, S.; Rui, R.; Zhang, D.; Dai, J. Involvement of PINK1/Parkin-mediated mitophagy in mitochondrial functional disruption under oxidative stress in vitrified porcine oocytes. Theriogenology 2021, 174, 160–168. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, H.; Li, C.; Liu, Q.; Li, J.; Wu, H.; Xu, R.; Sun, Y.; Cheng, M.; Zhao, X.; Pan, M.; et al. C-type natriuretic peptide improves maternally aged oocytes quality by inhibiting excessive PINK1/Parkin-mediated mitophagy. Elife 2023, 12, RP88523. [Google Scholar] [CrossRef]
  115. Showell, M.G.; Mackenzie-Proctor, R.; Jordan, V.; Hart, R.J. Antioxidants for female subfertility. Cochrane Database Syst. Rev. 2020, 8, Cd007807. [Google Scholar] [CrossRef]
  116. Fang, X.; Xia, W.; Qi, Y.; Yu, Y.; Sun, Q.; Zhang, D.; Zhou, Z.; Qin, T.; Tao, C.; Li, J. SIRT2 regulates apoptosis by inducing mitophagy in sheep cumulus cells. Theriogenology 2024, 218, 163–173. [Google Scholar] [CrossRef]
  117. Sun, Y.L.; Tang, S.B.; Shen, W.; Yin, S.; Sun, Q.Y. Roles of Resveratrol in Improving the Quality of Postovulatory Aging Oocytes In Vitro. Cells 2019, 8, 1132. [Google Scholar] [CrossRef]
  118. Fang, X.; Xia, W.; Li, S.; Qi, Y.; Liu, M.; Yu, Y.; Li, H.; Li, M.; Tao, C.; Wang, Z.; et al. SIRT2 Is Critical for Sheep Oocyte Maturation through Regulating Function of Surrounding Granulosa Cells. Int. J. Mol. Sci. 2022, 23, 5013. [Google Scholar] [CrossRef]
  119. Shin, J.-H.; Yang, S.-G.; Hyo-Jin, P.; Koo, D.-B. Oocyte quality is closely linked to DRP1 derived-mitochondrial fission and mitophagy by the NAD+ biosynthesis in a postovulatory-aging model of pigs. J. Anim. Reprod. Biotechnol. 2024, 39, 67–80. [Google Scholar] [CrossRef]
  120. Ng, A.Q.E.; Chan, S.N.; Pek, J.W. Nutrient-dependent regulation of a stable intron modulates germline mitochondrial quality control. Nat. Commun. 2024, 15, 1252. [Google Scholar] [CrossRef]
  121. Wong, J.T.; Akhbar, F.; Ng, A.Y.E.; Tay, M.L.; Loi, G.J.E.; Pek, J.W. DIP1 modulates stem cell homeostasis in Drosophila through regulation of sisR-1. Nat. Commun. 2017, 8, 759. [Google Scholar] [CrossRef] [PubMed]
  122. Jin, X.; Wang, K.; Wang, L.; Liu, W.; Zhang, C.; Qiu, Y.; Liu, W.; Zhang, H.; Zhang, D.; Yang, Z.; et al. RAB7 activity is required for the regulation of mitophagy in oocyte meiosis and oocyte quality control during ovarian aging. Autophagy 2022, 18, 643–660. [Google Scholar] [CrossRef]
  123. Yan, B.R.; Li, T.; Coyaud, E.; Laurent, E.M.N.; St-Germain, J.; Zhou, Y.; Kim, P.K.; Raught, B.; Brumell, J.H. C5orf51 is a component of the MON1-CCZ1 complex and controls RAB7A localization and stability during mitophagy. Autophagy 2022, 18, 829–840. [Google Scholar] [CrossRef] [PubMed]
  124. Ma, Q.; Shen, M.; Wu, J.; Ye, C.; Tan, Y. Mechanism Research of DHEA Treatment Improving Diminished Ovarian Reserve by Attenuating the AMPK-SIRT1 Signaling and Mitophagy. Reprod. Sci. 2024, 31, 2059–2072. [Google Scholar] [CrossRef]
  125. Furat Rencber, S.; Kurnaz Ozbek, S.; Eraldemır, C.; Sezer, Z.; Kum, T.; Ceylan, S.; Guzel, E. Effect of resveratrol and metformin on ovarian reserve and ultrastructure in PCOS: An experimental study. J. Ovarian Res. 2018, 11, 55. [Google Scholar] [CrossRef]
  126. Zhang, Y.; Bai, J.; Cui, Z.; Li, Y.; Gao, Q.; Miao, Y.; Xiong, B. Polyamine metabolite spermidine rejuvenates oocyte quality by enhancing mitophagy during female reproductive aging. Nat. Aging 2023, 3, 1372–1386. [Google Scholar] [CrossRef]
  127. Field, J.T.; Gordon, J.W. BNIP3 and Nix: Atypical regulators of cell fate. Biochim. Biophys. Acta Mol. Cell Res. 2022, 1869, 119325. [Google Scholar] [CrossRef]
  128. Li, Y.; Zheng, W.; Lu, Y.; Zheng, Y.; Pan, L.; Wu, X.; Yuan, Y.; Shen, Z.; Ma, S.; Zhang, X.; et al. BNIP3L/NIX-mediated mitophagy: Molecular mechanisms and implications for human disease. Cell Death Dis. 2021, 13, 14. [Google Scholar] [CrossRef]
  129. Palikaras, K.; Lionaki, E.; Tavernarakis, N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat. Cell Biol. 2018, 20, 1013–1022. [Google Scholar] [CrossRef]
  130. Liu, L.; Feng, D.; Chen, G.; Chen, M.; Zheng, Q.; Song, P.; Ma, Q.; Zhu, C.; Wang, R.; Qi, W.; et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat. Cell Biol. 2012, 14, 177–185. [Google Scholar] [CrossRef]
  131. Chen, M.; Chen, Z.; Wang, Y.; Tan, Z.; Zhu, C.; Li, Y.; Han, Z.; Chen, L.; Gao, R.; Liu, L.; et al. Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy. Autophagy 2016, 12, 689–702. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, J.; Zhu, P.; Li, R.; Ren, J.; Zhou, H. Fundc1-dependent mitophagy is obligatory to ischemic preconditioning-conferred renoprotection in ischemic AKI via suppression of Drp1-mediated mitochondrial fission. Redox Biol. 2020, 30, 101415. [Google Scholar] [CrossRef] [PubMed]
  133. Wu, W.; Li, W.; Chen, H.; Jiang, L.; Zhu, R.; Feng, D. FUNDC1 is a novel mitochondrial-associated-membrane (MAM) protein required for hypoxia-induced mitochondrial fission and mitophagy. Autophagy 2016, 12, 1675–1676. [Google Scholar] [CrossRef] [PubMed]
  134. Jia, L.; Yang, Y.; Sun, F.; Tao, H.; Lu, C.; Yang, J.J. Mitochondrial quality control in liver fibrosis: Epigenetic hallmarks and therapeutic strategies. Cell. Signal. 2024, 115, 111035. [Google Scholar] [CrossRef]
  135. Lin, L.C.; Tu, B.; Song, K.; Liu, Z.Y.; Sun, H.; Zhou, Y.; Sha, J.M.; Yang, J.J.; Zhang, Y.; Zhao, J.Y.; et al. Mitochondrial quality control in cardiac fibrosis: Epigenetic mechanisms and therapeutic strategies. Metabolism 2023, 145, 155626. [Google Scholar] [CrossRef]
  136. Feng, Y.; Lu, Y. The nuclear-mitochondrial crosstalk in aging: From mechanisms to therapeutics. Free Radic. Biol. Med. 2025, 232, 391–397. [Google Scholar] [CrossRef]
  137. Naderi, N.; Tavalaee, M.; Nasr-Esfahani, M.H. The epigenetic approach of varicocele: A focus on sperm DNA and m6A-RNA methylation. Hum. Reprod. Update 2025, 31, 81–101. [Google Scholar] [CrossRef]
  138. Balasubramanian, N.; Jadhav, G.; Sakharkar, A.J. Repeated mild traumatic brain injuries perturb the mitochondrial biogenesis via DNA methylation in the hippocampus of rat. Mitochondrion 2021, 61, 11–24. [Google Scholar] [CrossRef]
  139. Balasubramanian, N.; Sagarkar, S.; Choudhary, A.G.; Kokare, D.M.; Sakharkar, A.J. Epigenetic Blockade of Hippocampal SOD2 Via DNMT3b-Mediated DNA Methylation: Implications in Mild Traumatic Brain Injury-Induced Persistent Oxidative Damage. Mol. Neurobiol. 2021, 58, 1162–1184. [Google Scholar] [CrossRef]
  140. Janssen, B.G.; Byun, H.M.; Gyselaers, W.; Lefebvre, W.; Baccarelli, A.A.; Nawrot, T.S. Placental mitochondrial methylation and exposure to airborne particulate matter in the early life environment: An ENVIRONAGE birth cohort study. Epigenetics 2015, 10, 536–544. [Google Scholar] [CrossRef]
  141. Janssen, B.G.; Byun, H.M.; Roels, H.A.; Gyselaers, W.; Penders, J.; Baccarelli, A.A.; Nawrot, T.S. Regulating role of fetal thyroid hormones on placental mitochondrial DNA methylation: Epidemiological evidence from the ENVIRONAGE birth cohort study. Clin. Epigenet. 2017, 9, 66. [Google Scholar] [CrossRef] [PubMed]
  142. He, S.; Qu, Q.; Chen, X.; Zhao, L.; Jiao, Z.; Wan, Z.; Kwok, H.F.; Qu, S. Downregulation of Ambra1 by altered DNA methylation exacerbates dopaminergic neuron damage in a fenpropathrin-induced Parkinson-like mouse model. Ecotoxicol. Environ. Saf. 2024, 271, 115995. [Google Scholar] [CrossRef] [PubMed]
  143. Zhou, J.; Zhao, Y.; Li, Z.; Zhu, M.; Wang, Z.; Li, Y.; Xu, T.; Feng, D.; Zhang, S.; Tang, F.; et al. miR-103a-3p regulates mitophagy in Parkinson’s disease through Parkin/Ambra1 signaling. Pharmacol. Res. 2020, 160, 105197. [Google Scholar] [CrossRef] [PubMed]
  144. Deng, Z.; Yao, J.; Xiao, N.; Han, Y.; Wu, X.; Ci, C.; Chen, K.; Geng, X. DNA methyltransferase 1 (DNMT1) suppresses mitophagy and aggravates heart failure via the microRNA-152-3p/ETS1/RhoH axis. Lab. Investig. 2022, 102, 782–793. [Google Scholar] [CrossRef]
  145. Jin, F.; Li, M.; Li, X.; Zheng, Y.; Zhang, K.; Liu, X.; Cai, B.; Yin, G. DNMT1-mediated methylation inhibits microRNA-214-3p and promotes hair follicle stem cell differentiate into adipogenic lineages. Stem Cell Res. Ther. 2020, 11, 444. [Google Scholar] [CrossRef]
  146. Jiang, S.X.; Zhou, Z.Y.; Tu, B.; Song, K.; Lin, L.C.; Liu, Z.Y.; Cao, W.; Zhao, J.Y.; Tao, H. Epigenetic regulation of mitochondrial fission and cardiac fibrosis via sFRP3 promoter methylation. Cell. Mol. Life Sci. 2024, 81, 483. [Google Scholar] [CrossRef]
  147. Jia, Y.; Xie, H.; Wu, S.; Dong, J.; Ying, H. Induction of FAM46C expression mediated by DNMT3A downregulation is involved in early-onset preeclampsia through gene body methylation. Cell. Signal. 2025, 125, 111506. [Google Scholar] [CrossRef]
  148. Li, X.C.; Jin, F.; Wang, B.Y.; Yin, X.J.; Hong, W.; Tian, F.J. The m6A demethylase ALKBH5 controls trophoblast invasion at the maternal-fetal interface by regulating the stability of CYR61 mRNA. Theranostics 2019, 9, 3853–3865. [Google Scholar] [CrossRef]
  149. Cotney, J.; McKay, S.E.; Shadel, G.S. Elucidation of separate, but collaborative functions of the rRNA methyltransferase-related human mitochondrial transcription factors B1 and B2 in mitochondrial biogenesis reveals new insight into maternally inherited deafness. Hum. Mol. Genet. 2009, 18, 2670–2682. [Google Scholar] [CrossRef]
  150. Raimundo, N.; Song, L.; Shutt, T.E.; McKay, S.E.; Cotney, J.; Guan, M.X.; Gilliland, T.C.; Hohuan, D.; Santos-Sacchi, J.; Shadel, G.S. Mitochondrial stress engages E2F1 apoptotic signaling to cause deafness. Cell 2012, 148, 716–726. [Google Scholar] [CrossRef]
  151. Kahl, M.; Xu, Z.; Arumugam, S.; Edens, B.; Fischietti, M.; Zhu, A.C.; Platanias, L.C.; He, C.; Zhuang, X.; Ma, Y.C. m6A RNA methylation regulates mitochondrial function. Hum. Mol. Genet. 2024, 33, 969–980. [Google Scholar] [CrossRef] [PubMed]
  152. Zhu, Y.; Yang, B.; Chen, S.; Chen, G.; Zeng, X.; Min, H.; Xu, L. M6A RNA Methylation-Mediated TUG1 Stability Maintains Mitochondrial Homeostasis during Kidney Aging by Epigenetically Regulating PGC1-α Expression. Antioxid. Redox Signal. 2024, 41, 993–1013. [Google Scholar] [CrossRef] [PubMed]
  153. Tu, B.; Song, K.; Zhou, Y.; Sun, H.; Liu, Z.Y.; Lin, L.C.; Ding, J.F.; Sha, J.M.; Shi, Y.; Yang, J.J.; et al. METTL3 boosts mitochondrial fission and induces cardiac fibrosis by enhancing LncRNA GAS5 methylation. Pharmacol. Res. 2023, 194, 106840. [Google Scholar] [CrossRef]
  154. Zhou, Y.; Song, K.; Tu, B.; Sun, H.; Ding, J.F.; Luo, Y.; Sha, J.M.; Li, R.; Zhang, Y.; Zhao, J.Y.; et al. METTL3 boosts glycolysis and cardiac fibroblast proliferation by increasing AR methylation. Int. J. Biol. Macromol. 2022, 223, 899–915. [Google Scholar] [CrossRef]
  155. Zhou, J.; Zhang, H.; Zhong, K.; Tao, L.; Lin, Y.; Xie, G.; Tan, Y.; Wu, Y.; Lu, Y.; Chen, Z.; et al. N6-methyladenosine facilitates mitochondrial fusion of colorectal cancer cells via induction of GSH synthesis and stabilization of OPA1 mRNA. Natl. Sci. Rev. 2024, 11, nwae039. [Google Scholar] [CrossRef]
  156. Shen, C.; Xuan, B.; Yan, T.; Ma, Y.; Xu, P.; Tian, X.; Zhang, X.; Cao, Y.; Ma, D.; Zhu, X.; et al. m(6)A-dependent glycolysis enhances colorectal cancer progression. Mol. Cancer 2020, 19, 72. [Google Scholar] [CrossRef]
  157. Li, J.; Zhang, S.; Li, C.; Zhang, X.; Shan, Y.; Zhang, Z.; Bo, H.; Zhang, Y. Endurance exercise-induced histone methylation modification involved in skeletal muscle fiber type transition and mitochondrial biogenesis. Sci. Rep. 2024, 14, 21154. [Google Scholar] [CrossRef]
  158. Li, J.; Zhang, Z.; Bo, H.; Zhang, Y. Exercise couples mitochondrial function with skeletal muscle fiber type via ROS-mediated epigenetic modification. Free Radic. Biol. Med. 2024, 213, 409–425. [Google Scholar] [CrossRef]
  159. Zhang, Y.; Wang, X.; Mu, Q.; Hou, X.; Yu, W.; Guo, J. Histone H3 Acetylation Is Involved in Retinoid Acid-Induced Neural Differentiation through Increasing Mitochondrial Function. Biomedicines 2023, 11, 3251. [Google Scholar] [CrossRef]
  160. Wiesel-Motiuk, N.; Assaraf, Y.G. The key roles of the lysine acetyltransferases KAT6A and KAT6B in physiology and pathology. Drug Resist. Updates 2020, 53, 100729. [Google Scholar] [CrossRef]
  161. Liu, X.; Chen, Y.; Zhao, L.; Tian, Q.; deAvila, J.M.; Zhu, M.J.; Du, M. Dietary succinate supplementation to maternal mice improves fetal brown adipose tissue development and thermogenesis of female offspring. J. Nutr. Biochem. 2022, 100, 108908. [Google Scholar] [CrossRef] [PubMed]
  162. Wu, W.; Jia, H.; Chen, S.; Ma, X.; Zhou, S.; Qiu, L.; Wu, X.; Li, P.; Chu, H.; Zhang, G. Inhibition of OGG1 ameliorates pulmonary fibrosis via preventing M2 macrophage polarization and activating PINK1-mediated mitophagy. Mol. Med. 2024, 30, 72. [Google Scholar] [CrossRef] [PubMed]
  163. Zhu, W.; Wang, C.; Xue, L.; Liu, L.; Yang, X.; Liu, Z.; Zhang, S.; Luo, D. The SMYD3-MTHFD1L-formate metabolic regulatory axis mediates mitophagy to inhibit M1 polarization in macrophages. Int. Immunopharmacol. 2022, 113, 109352. [Google Scholar] [CrossRef]
  164. Zhang, X.; Zhang, L.; Xiang, W. The impact of mitochondrial dysfunction on ovarian aging. J. Transl. Med. 2025, 23, 211. [Google Scholar] [CrossRef]
  165. Huang, F.; Luo, X.; Ou, Y.; Gao, Z.; Tang, Q.; Chu, Z.; Zhu, X.; He, Y. Control of histone demethylation by nuclear-localized α-ketoglutarate dehydrogenase. Science 2023, 381, eadf8822. [Google Scholar] [CrossRef]
  166. Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef]
  167. Mizuarai, S.; Miki, S.; Araki, H.; Takahashi, K.; Kotani, H. Identification of dicarboxylate carrier Slc25a10 as malate transporter in de novo fatty acid synthesis. J. Biol. Chem. 2005, 280, 32434–32441. [Google Scholar] [CrossRef]
  168. Yuan, H.; Wu, X.; Wu, Q.; Chatoff, A.; Megill, E.; Gao, J.; Huang, T.; Duan, T.; Yang, K.; Jin, C.; et al. Lysine catabolism reprograms tumour immunity through histone crotonylation. Nature 2023, 617, 818–826. [Google Scholar] [CrossRef]
  169. Zhu, D.; Li, X.; Tian, Y. Mitochondrial-to-nuclear communication in aging: An epigenetic perspective. Trends Biochem. Sci. 2022, 47, 645–659. [Google Scholar] [CrossRef]
  170. Trefely, S.; Lovell, C.D.; Snyder, N.W.; Wellen, K.E. Compartmentalised acyl-CoA metabolism and roles in chromatin regulation. Mol. Metab. 2020, 38, 100941. [Google Scholar] [CrossRef]
  171. Hao, X.; Kuang, C.F.; Gu, Z.T.; Wang, Y.F.; Li, S.A.; Ku, Y.L.; Li, Y.H.; Ge, J.H.; Liu, X. From microscopy to nanoscopy via visible light. Light-Sci. Appl. 2013, 2, e108. [Google Scholar] [CrossRef]
  172. Bialas, N.; Sokolova, V.; van der Meer, S.B.; Knuschke, T.; Ruks, T.; Klein, K.; Westendorf, A.M.; Epple, M. Bacteria (E. coli) take up ultrasmall gold nanoparticles (2 nm) as shown by different optical microscopic techniques (CLSM, SIM, STORM). Nano Select 2022, 3, 1407–1420. [Google Scholar] [CrossRef]
  173. Frolikova, M.; Blazikova, M.; Capek, M.; Chmelova, H.; Valecka, J.; Kolackova, V.; Valaskova, E.; Gregor, M.; Komrskova, K.; Horvath, O.; et al. Innovative sample preparation using alcohol dehydration and high refractive index medium enables acquisition of two-channel super-resolution 3D STED image of an entire oocyte. J. Microsc. 2025, 297, 165–178. [Google Scholar] [CrossRef] [PubMed]
  174. Wei, Y.; He, X.; Fang, J.B.; Ju, M.Z. Spatiotemporal dynamic monitoring of protein conformation changes by super-resolution imaging. Dye. Pigment. 2025, 236, 112689. [Google Scholar] [CrossRef]
  175. Yang, X.Z.; Fang, H.B.; Li, S.M.; Chu, C.Y.; Zhang, Y.H.; Yang, Y.; He, W.J.; Chen, Y.C.; Guo, Z.J. Aggregation-based dual-target probe for dual-colour super-resolution monitoring mitophagy and evaluating drugs regulating mitochondria. Aggregate 2025, 6, e641. [Google Scholar] [CrossRef]
  176. Gao, S.; Sun, J.; Hou, Y.; Ge, X.; Shi, M.; Zheng, H.; Zhang, Y.; Li, M.; Gao, B.; Xi, P. HBimmCue: A Versatile Fluorescent Probe for Multi-Scale Imaging of Lipid Polarity and Membrane Order in Inner Mitochondrial Membrane. Adv. Sci. 2025, 12, e2414343. [Google Scholar] [CrossRef]
  177. Vargas-Ordaz, E.; Newman, H.; Austin, C.; Catt, S.; Nosrati, R.; Cadarso, V.J.; Neild, A.; Horta, F. Novel application of metabolic imaging of early embryos using a light-sheet on-a-chip device: A proof-of-concept study. Hum. Reprod. 2025, 40, 41–55. [Google Scholar] [CrossRef]
  178. Chakraborty, C.; Bhattacharya, M.; Agoramoorthy, G. Single-cell sequencing of miRNAs: A modified technology. Cell Biol. Int. 2020, 44, 1773–1780. [Google Scholar] [CrossRef]
  179. Song, K.; Yang, X.; An, G.; Xia, X.; Zhao, J.; Xu, X.; Wan, C.; Liu, T.; Zheng, Y.; Ren, S.; et al. Targeting APLN/APJ restores blood-testis barrier and improves spermatogenesis in murine and human diabetic models. Nat. Commun. 2022, 13, 7335. [Google Scholar] [CrossRef]
  180. Liu, C.; Zuo, W.; Yan, G.; Wang, S.; Sun, S.; Li, S.; Tang, X.; Li, Y.; Cai, C.; Wang, H.; et al. Granulosa cell mevalonate pathway abnormalities contribute to oocyte meiotic defects and aneuploidy. Nat. Aging 2023, 3, 670–687. [Google Scholar] [CrossRef]
  181. Gao, L.; Zhang, C.; Zheng, Y.; Wu, D.; Chen, X.; Lan, H.; Zheng, X.; Wu, H.; Li, S. Glycine regulates lipid peroxidation promoting porcine oocyte maturation and early embryonic development. J. Anim. Sci. 2023, 101, skac425. [Google Scholar] [CrossRef]
  182. Hashimshony, T.; Senderovich, N.; Avital, G.; Klochendler, A.; de Leeuw, Y.; Anavy, L.; Gennert, D.; Li, S.; Livak, K.J.; Rozenblatt-Rosen, O.; et al. CEL-Seq2: Sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 2016, 17, 77. [Google Scholar] [CrossRef]
  183. Hashimshony, T.; Wagner, F.; Sher, N.; Yanai, I. CEL-Seq: Single-cell RNA-Seq by multiplexed linear amplification. Cell Rep. 2012, 2, 666–673. [Google Scholar] [CrossRef]
  184. Sasagawa, Y.; Nikaido, I.; Hayashi, T.; Danno, H.; Uno, K.D.; Imai, T.; Ueda, H.R. Quartz-Seq: A highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity. Genome Biol. 2013, 14, R31. [Google Scholar] [CrossRef]
  185. Homberger, C.; Imdahl, F.; Hayward, R.J.; Barquist, L.; Saliba, A.E.; Vogel, J. Transcriptomic profiling of individual bacteria by MATQ-seq. Nat. Protoc. 2025, 1–30. [Google Scholar] [CrossRef]
  186. Sheng, K.; Cao, W.; Niu, Y.; Deng, Q.; Zong, C. Effective detection of variation in single-cell transcriptomes using MATQ-seq. Nat. Methods 2017, 14, 267–270. [Google Scholar] [CrossRef]
  187. Sun, F.; Li, H.; Sun, D.; Fu, S.; Gu, L.; Shao, X.; Wang, Q.; Dong, X.; Duan, B.; Xing, F.; et al. Single-cell omics: Experimental workflow, data analyses and applications. Sci. China Life Sci. 2025, 68, 5–102. [Google Scholar] [CrossRef]
  188. Love, A.M.A.; Edwards, C.; Cai, R.Y.; Gibbs, V. Using Experience Sampling Methodology to Capture Disclosure Opportunities for Autistic Adults. Autism Adulthood 2023, 5, 389–400. [Google Scholar] [CrossRef]
  189. Huang, J.; Chen, P.; Jia, L.; Li, T.; Yang, X.; Liang, Q.; Zeng, Y.; Liu, J.; Wu, T.; Hu, W.; et al. Multi-Omics Analysis Reveals Translational Landscapes and Regulations in Mouse and Human Oocyte Aging. Adv. Sci. 2023, 10, e2301538. [Google Scholar] [CrossRef]
  190. Wu, Y.W.; Li, S.; Zheng, W.; Li, Y.C.; Chen, L.; Zhou, Y.; Deng, Z.Q.; Lin, G.; Fan, H.Y.; Sha, Q.Q. Dynamic mRNA degradome analyses indicate a role of histone H3K4 trimethylation in association with meiosis-coupled mRNA decay in oocyte aging. Nat. Commun. 2022, 13, 3191. [Google Scholar] [CrossRef]
  191. Zeng, C.; Wang, Y. Exploring the Cancer Immune Epigenome by ATAC-Seq. In Cancer Immunosurveillance. Methods in Molecular Biology; Humana: New York, NY, USA, 2025; Volume 2930, pp. 219–227. [Google Scholar] [CrossRef]
  192. Ming, H.; Sun, J.; Pasquariello, R.; Gatenby, L.; Herrick, J.R.; Yuan, Y.; Pinto, C.R.; Bondioli, K.R.; Krisher, R.L.; Jiang, Z. The landscape of accessible chromatin in bovine oocytes and early embryos. Epigenetics 2021, 16, 300–312. [Google Scholar] [CrossRef] [PubMed]
  193. Gou, L.T.; Lim, D.H.; Ma, W.; Aubol, B.E.; Hao, Y.; Wang, X.; Zhao, J.; Liang, Z.; Shao, C.; Zhang, X.; et al. Initiation of Parental Genome Reprogramming in Fertilized Oocyte by Splicing Kinase SRPK1-Catalyzed Protamine Phosphorylation. Cell 2020, 180, 1212–1227.e1214. [Google Scholar] [CrossRef] [PubMed]
  194. Wu, D.; Dean, J. Reduced female fertility due to sequestration of RNA Pol II by pervasive transcription in exosome RNase-depleted oocytes. Cell Rep. 2023, 42, 113247. [Google Scholar] [CrossRef] [PubMed]
  195. Zhou, C.; Halstead, M.M.; Bonnet-Garnier, A.; Schultz, R.M.; Ross, P.J. Histone remodeling reflects conserved mechanisms of bovine and human preimplantation development. EMBO Rep. 2023, 24, e55726. [Google Scholar] [CrossRef]
  196. Zhang, B.; Qi, T.; Lin, J.; Zhai, S.; Wang, X.; Zhou, L.; Deng, X. KLF6-mediated recruitment of the p300 complex enhances H3K23su and cooperatively upregulates SEMA3C with FOSL2 to drive 5-FU resistance in colon cancer cells. Exp. Mol. Med. 2025, 57, 667–685. [Google Scholar] [CrossRef]
  197. Zhao, X.M.; Wang, N.; Hao, H.S.; Li, C.Y.; Zhao, Y.H.; Yan, C.L.; Wang, H.Y.; Du, W.H.; Wang, D.; Liu, Y.; et al. Melatonin improves the fertilization capacity and developmental ability of bovine oocytes by regulating cytoplasmic maturation events. J. Pineal Res. 2018, 64, e12445. [Google Scholar] [CrossRef]
  198. Gu, J.; Hua, R.; Wu, H.; Guo, C.; Hai, Z.; Xiao, Y.; Yeung, W.S.B.; Liu, K.; Babayev, E.; Wang, T. Salidroside Improves Oocyte Competence of Reproductively Old Mice by Enhancing Mitophagy. Aging Cell 2025, e14475. [Google Scholar] [CrossRef]
  199. Leathersich, S.J.; Roche, C.S.; Walls, M.; Nathan, E.; Hart, R.J. Particulate air pollution at the time of oocyte retrieval is independently associated with reduced odds of live birth in subsequent frozen embryo transfers. Hum. Reprod. 2025, 40, 110–118. [Google Scholar] [CrossRef]
  200. Erdenetogtokh, P.; Kanno, C.; Sakaguchi, K.; Yanagawa, Y.; Katagiri, S.; Nagano, M. Effect of astaxanthin addition to an individual culture system for in vitro maturation of bovine oocytes on accumulation of reactive oxygen species and mitochondrial activity. Jpn. J. Vet. Res. 2018, 66, 325–329. [Google Scholar] [CrossRef]
  201. Khan, A.M.; Idrees, M.; Perera, C.D.; Haider, Z.; Joo, M.D.; Kang, J.S.; Lee, S.H.; Kong, I.K. The effects of cycloastragenol on bovine embryo development, implantation potential and telomerase activity. Reprod. Fertil. Dev. 2023, 35, 527–538. [Google Scholar] [CrossRef]
  202. Gutierrez-Castillo, E.; Diaz, F.A.; Talbot, S.A.; Bondioli, K.R. Effect of bovine oocyte vitrification with EGTA and post-warming recovery with resveratrol on meiotic spindle, mitochondrial function, reactive oxygen species, and developmental competence. Theriogenology 2023, 196, 59–67. [Google Scholar] [CrossRef] [PubMed]
  203. Gutierrez-Castillo, E.; Diaz, F.A.; Talbot, S.A.; Bondioli, K.R. Recovery of spindle morphology and mitochondrial function through extended culture after vitrification-warming of bovine oocytes. Theriogenology 2022, 189, 192–198. [Google Scholar] [CrossRef] [PubMed]
  204. Chinen, S.; Yamanaka, T.; Hirabayashi, M.; Hochi, S. Rescue of vitrified-warmed bovine mature oocytes by short-term recovery culture with resveratrol. Cryobiology 2020, 97, 185–190. [Google Scholar] [CrossRef]
  205. Sprícigo, J.F.; Morató, R.; Arcarons, N.; Yeste, M.; Dode, M.A.; López-Bejar, M.; Mogas, T. Assessment of the effect of adding L-carnitine and/or resveratrol to maturation medium before vitrification on in vitro-matured calf oocytes. Theriogenology 2017, 89, 47–57. [Google Scholar] [CrossRef]
  206. Ito, J.; Iwata, H. Age-related advanced glycation end-product accumulation impairs mitochondrial regulation after vitrification†. Biol. Reprod. 2023, 109, 271–281. [Google Scholar] [CrossRef]
  207. Yang, C.X.; Liu, S.; Miao, J.K.; Mou, Q.; Liu, X.M.; Wang, P.C.; Huo, L.J.; Du, Z.Q. CoQ10 improves meiotic maturation of pig oocytes through enhancing mitochondrial function and suppressing oxidative stress. Theriogenology 2021, 159, 77–86. [Google Scholar] [CrossRef]
  208. He, X.; Chen, H.; Liao, M.; Zhao, X.; Zhang, D.; Jiang, M.; Jiang, Z. The role of CoQ10 in embryonic development. J. Assist. Reprod. Genet. 2024, 41, 767–779. [Google Scholar] [CrossRef]
  209. Ratchamak, R.; Authaida, S.; Koedkanmark, T.; Boonkum, W.; Chankitisakul, V. Coenzyme Q10 Supplementation Effects on In Vitro Oocyte Maturation, Lipid Peroxidation, and Embryonic Development in Prepubertal and Aging Thai-Holstein Cows. Animals 2024, 15, 18. [Google Scholar] [CrossRef]
  210. Bisogno, S.; Depciuch, J.; Gulzar, H.; Heber, M.F.; Kobiałka, M.; Gąsior, Ł.; Bereta, A.; Pieczara, A.; Fic, K.; Musson, R.; et al. Female-age-dependent changes in the lipid fingerprint of the mammalian oocytes. Hum. Reprod. 2024, 39, 2754–2767. [Google Scholar] [CrossRef]
  211. Laplacette, A.L.; Rial, C.; Sitko, E.; Perez, M.M.; Tompkins, S.; Stangaferro, M.L.; Thomas, M.J.; Giordano, J.O. Delaying induction of ovulation and timed artificial insemination in a Double-Ovsynch protocol increased expression of estrus and altered first-service reproductive outcomes of lactating dairy cows. J. Dairy Sci. 2025, 108, 1103–1124. [Google Scholar] [CrossRef]
  212. Ramírez-Martín, N.; Buigues, A.; Rodríguez-Varela, C.; Martínez, J.; Blázquez-Simón, P.; Rodríguez-Hernández, C.; Pellicer, N.; Pellicer, A.; Escribá, M.J.; Herraiz, S. Nicotinamide mononucleotide supplementation improves oocyte developmental competence in different ovarian damage conditions. Am. J. Obstet. Gynecol. 2025. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, H.; Qin, X.; Bojan, N.; Cao, C.; Chai, J.; Pang, W. Nicotinamide mononucleotide enhances porcine sperm quality by activating the SIRT3-SOD2/ROS pathway and promoting oxidative phosphorylation. Anim. Reprod. Sci. 2025, 275, 107797. [Google Scholar] [CrossRef] [PubMed]
  214. Li, H.J.; Sutton-McDowall, M.L.; Wang, X.; Sugimura, S.; Thompson, J.G.; Gilchrist, R.B. Extending prematuration with cAMP modulators enhances the cumulus contribution to oocyte antioxidant defence and oocyte quality via gap junctions. Hum. Reprod. 2016, 31, 810–821. [Google Scholar] [CrossRef] [PubMed]
  215. An, Q.; Peng, W.; Cheng, Y.; Lu, Z.; Zhou, C.; Zhang, Y.; Su, J. Melatonin supplementation during in vitro maturation of oocyte enhances subsequent development of bovine cloned embryos. J. Cell. Physiol. 2019, 234, 17370–17381. [Google Scholar] [CrossRef]
  216. El Sheikh, M.; Mesalam, A.; Mesalam, A.A.; Idrees, M.; Lee, K.L.; Kong, I.K. Melatonin Abrogates the Anti-Developmental Effect of the AKT Inhibitor SH6 in Bovine Oocytes and Embryos. Int. J. Mol. Sci. 2019, 20, 2956. [Google Scholar] [CrossRef]
  217. Santiquet, N.W.; Greene, A.F.; Becker, J.; Barfield, J.P.; Schoolcraft, W.B.; Krisher, R.L. A pre-in vitro maturation medium containing cumulus oocyte complex ligand-receptor signaling molecules maintains meiotic arrest, supports the cumulus oocyte complex and improves oocyte developmental competence. Mol. Hum. Reprod. 2017, 23, 594–606. [Google Scholar] [CrossRef]
  218. Truong, T.T.; Soh, Y.M.; Gardner, D.K. Antioxidants improve mouse preimplantation embryo development and viability. Hum. Reprod. 2016, 31, 1445–1454. [Google Scholar] [CrossRef]
  219. Novielli, C.; Anelli, G.M.; Lisso, F.; Marzorati, A.; Parrilla, B.; Oneta, M.; Savasi, V.M.; Cetin, I.; Mandò, C. Effects of α-lipoic acid and myo-inositol supplementation on the oocyte environment of infertile obese women: A preliminary study. Reprod. Biol. 2020, 20, 541–546. [Google Scholar] [CrossRef]
  220. Silva, B.R.; Silva, J.R.V. Mechanisms of action of non-enzymatic antioxidants to control oxidative stress during in vitro follicle growth, oocyte maturation, and embryo development. Anim. Reprod. Sci. 2023, 249, 107186. [Google Scholar] [CrossRef]
  221. Jiang, W.; Li, Y.; Zhao, Y.; Gao, Q.; Jin, Q.; Yan, C.; Xu, Y. l-carnitine supplementation during in vitro culture regulates oxidative stress in embryos from bovine aged oocytes. Theriogenology 2020, 143, 64–73. [Google Scholar] [CrossRef]
Figure 1. Mitochondrial essential functions in oocyte maturation: encompassing bioenergetic supply, coordinated calcium regulation, ROS dynamics, cellular signaling, and apoptotic control. Up arrow: Upregulating; Down arrow: Downregulating. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Figure 1. Mitochondrial essential functions in oocyte maturation: encompassing bioenergetic supply, coordinated calcium regulation, ROS dynamics, cellular signaling, and apoptotic control. Up arrow: Upregulating; Down arrow: Downregulating. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Animals 15 02000 g001
Figure 2. Mitochondrial quality control pathways in oocytes. Mitochondrial biogenesis, mitochondrial dynamics, and mitophagy are closely interrelated. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Figure 2. Mitochondrial quality control pathways in oocytes. Mitochondrial biogenesis, mitochondrial dynamics, and mitophagy are closely interrelated. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Animals 15 02000 g002
Figure 3. The major signaling pathways for mitochondrial biogenesis. PGC-1α and NRF1/2 serve as core transcriptional regulators that orchestrate mitochondrial DNA replication and electron transport chain assembly through transcriptional control of TFAM. Up arrow: Upregulating Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Figure 3. The major signaling pathways for mitochondrial biogenesis. PGC-1α and NRF1/2 serve as core transcriptional regulators that orchestrate mitochondrial DNA replication and electron transport chain assembly through transcriptional control of TFAM. Up arrow: Upregulating Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Animals 15 02000 g003
Figure 4. Mitochondrial dynamics: core regulatory machinery and modulatory determinants. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Figure 4. Mitochondrial dynamics: core regulatory machinery and modulatory determinants. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Animals 15 02000 g004
Figure 5. Mitophagy-regulated signaling pathways. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Figure 5. Mitophagy-regulated signaling pathways. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Animals 15 02000 g005
Figure 6. Mitochondria coordinate epigenetic modifications and quality control through metabolite-mediated regulation. Mitochondrial metabolites, such as α-KG, acetyl-CoA, NAD+, and succinate, serve as key substrates or cofactors that directly modify chromatin and histones. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Figure 6. Mitochondria coordinate epigenetic modifications and quality control through metabolite-mediated regulation. Mitochondrial metabolites, such as α-KG, acetyl-CoA, NAD+, and succinate, serve as key substrates or cofactors that directly modify chromatin and histones. Created in BioRender. https://www.biorender.com/ (accessed on 10 April 2025).
Animals 15 02000 g006
Table 1. Substances that improve mitochondrial function and oocyte quality.
Table 1. Substances that improve mitochondrial function and oocyte quality.
SubstancesFunctionReferences
ResveratrolMeiotic spindle stabilization; intracellular ROS reduction; mitochondrial biogenesis activation; cytoplasmic maturation enhancement; blastocyst formation promotion[107,197,202,203,204,205,206]
Coenzyme Q10Suppresses ROS generation; mitigates oxidative stress-induced apoptosis; facilitates nuclear maturation; enhances oocyte quality; improves embryonic developmental competence; alleviates oxidative stress; reinforces mitochondrial function; accelerates developmental progression; promotes blastocyst formation rates[207,208,209,210,211]
Nicotinamide MononucleotideROS accumulation attenuation; meiotic chromosomal misalignment correction; mitochondrial membrane potential restoration; ATP synthesis augmentation; mitochondrial autophagy activation; oocyte maturation rate elevation; spindle assembly fidelity preservation; NAD+ pool replenishment in cumulus–oocyte complexes[212,213]
MelatoninROS scavenging system activation (glutathione/antioxidant genes); mitochondrial architecture–function coordination; epigenetic regulation maintenance (methylation/hydroxymethylation)[197,214,215,216,217]
α Lipoic AcidMitochondrial functional boost (activity/mtDNA); transcriptional fine-tuning; oxidative damage neutralization[218,219]
L-carnitineMetabolic flux optimization (fatty acid/glucose/respiratory chain); oxidative stress–apoptosis axis suppression; oocyte rejuvenation triad (glutathione/membrane potential/cytoplasmic maturation)[220,221]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, Y.-R.; Xu, D.-J. Mitochondrial Quality Control in Bovine Oocyte Maturation: Mechanisms, Challenges, and Prospects for Enhancing Reproductive Efficiency. Animals 2025, 15, 2000. https://doi.org/10.3390/ani15132000

AMA Style

Zhang Y-R, Xu D-J. Mitochondrial Quality Control in Bovine Oocyte Maturation: Mechanisms, Challenges, and Prospects for Enhancing Reproductive Efficiency. Animals. 2025; 15(13):2000. https://doi.org/10.3390/ani15132000

Chicago/Turabian Style

Zhang, Yi-Ran, and De-Jun Xu. 2025. "Mitochondrial Quality Control in Bovine Oocyte Maturation: Mechanisms, Challenges, and Prospects for Enhancing Reproductive Efficiency" Animals 15, no. 13: 2000. https://doi.org/10.3390/ani15132000

APA Style

Zhang, Y.-R., & Xu, D.-J. (2025). Mitochondrial Quality Control in Bovine Oocyte Maturation: Mechanisms, Challenges, and Prospects for Enhancing Reproductive Efficiency. Animals, 15(13), 2000. https://doi.org/10.3390/ani15132000

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop