Next Article in Journal
Cytokine Receptor-like Factor 3 (CRLF3) and Its Emerging Roles in Neurobiology, Hematopoiesis and Related Human Diseases
Previous Article in Journal
Single-Cell RNA Sequencing of Baseline Immune Profiles After Third Vaccination Associated with Subsequent SARS-CoV-2 Infection in Naïve Individuals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities

Health Science Center, Hunan Normal University, Changsha 410013, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(8), 3496; https://doi.org/10.3390/ijms26083496
Submission received: 8 February 2025 / Revised: 1 April 2025 / Accepted: 4 April 2025 / Published: 8 April 2025
(This article belongs to the Special Issue Molecular Aspects of Reproductive Medicine)

Abstract

Male infertility is intricately linked to dysregulated cell death pathways, including ferroptosis, cuproptosis, pyroptosis, and autophagy. Ferroptosis, driven by iron-dependent lipid peroxidation through the Fenton reaction and inactivation of the GPX4/Nrf2/SLC7A11 axis, disrupts spermatogenesis under conditions of oxidative stress, environmental toxin exposure, or metabolic disorders. Similarly, cuproptosis—characterized by mitochondrial dysfunction and disulfide stress due to copper overload—exacerbates germ cell apoptosis via FDX1 activation and NADPH depletion. Pyroptosis, mediated by the NLRP3 inflammasome and gasdermin D, amplifies testicular inflammation and germ cell loss via IL-1β/IL-18 release, particularly in response to environmental insults. Autophagy maintains testicular homeostasis by clearing damaged organelles and proteins; however, its dysregulation impairs sperm maturation and compromises blood–testis barrier integrity. These pathways intersect through shared regulators; reactive oxygen species and mTOR modulate the autophagy–pyroptosis balance, while Nrf2 and FDX1 bridge ferroptosis–cuproptosis crosstalk. Therapeutic interventions targeting these mechanisms have shown promise in preclinical models. However, challenges persist, including the tissue-specific roles of gasdermin isoforms, off-target effects of pharmacological inhibitors, and transgenerational epigenetic impacts of environmental toxins. This review synthesizes current molecular insights into the cell death pathways implicated in male infertility, emphasizing their interplay and translational potential for restoring spermatogenic function.

1. Introduction

Male infertility is an increasingly recognized public health concern, with a global infertility rate of approximately 17.5% [1]. Systematic reviews and meta-analyses of retrospective cohort studies have demonstrated a significant decline in sperm counts among men worldwide from 1973 to 2018, encompassing regions including North America, Europe, Australia, South and Central America, Africa, and Asia [2]. Beyond its impact on reproductive health, severe male infertility is associated with an increased risk of chronic conditions, including cardiovascular diseases, autoimmune disorders, and malignant tumors such as testicular and prostate cancer [3]. Additionally, reduced sperm counts and concentrations have been associated with an increase in mortality rate [4,5]. A prospective cohort study of 384,419 Danish men revealed that infertile individuals exhibited a higher overall mortality rate compared to their fertile counterparts [6]. Furthermore, mortality risk demonstrates a dose-dependent relationship with the severity of semen quality impairment [7]. Therefore, male infertility constitutes a critical public health challenge, with significant implications for both individual reproductive health and population dynamics. Male infertility is multifactorial, influenced by environmental pollutants (e.g., endocrine disruptors), genetic anomalies (e.g., Y-chromosome microdeletions), congenital abnormalities (e.g., cryptorchidism), infections (e.g., SARS-CoV-2-induced pediatric acute epididymo-orchitis), lifestyle factors (e.g., smoking, obesity), and sociodemographic trends such as delayed parenthood [8,9,10,11,12].
The most prevalent etiology of male infertility, accounting for approximately 75% of cases, is impaired spermatogenesis secondary to primary testicular failure [13]. Spermatogenesis is a tightly regulated biological process wherein spermatogonial stem cells undergo self-renewal, differentiate through spermatocyte stages into spermatozoa, and ultimately mature into functionally competent sperm. This intricate process relies on a precisely coordinated testicular microenvironment comprising Sertoli cells, peritubular myoid cells, and Leydig cells, which collectively synthesize and secrete cytokines critical for spermatogenesis. Dysregulation of any cellular component with this microenvironment can lead to spermatogenic failure, consequently manifesting as male infertility [10]. Emerging evidence implicates regulated cell death pathways in the pathogenesis of male infertility (Figure 1). Distinct molecular mechanisms—including ferroptosis, cuproptosis, autophagy, and pyroptosis—have been identified as key mediators of germ cell demise, with activation patterns influenced by both the pathological state and environmental exposure. This review systematically examines the mechanistic interplay between these cell death modalities and male reproductive dysfunction, with particular emphasis on their pathophysiological significance and translational potential.

2. Ferroptosis

2.1. Male Infertility Is Closely Related to Ferroptosis

Ferroptosis, a form of cell death mediated by iron metabolism and lipid peroxidation, has been implicated in the pathogenesis of male infertility and impaired spermatogenesis. Clinical studies on male infertility or related comorbidities have shown abnormal levels of ferroptosis markers. Bioinformatic and machine learning studies suggest that the downregulation of heat shock transcription factor 1 (HSF1), Kelch domain-containing 3 (KLHDC3), and glutathione peroxidase 4 (GPX4), which is a selenium-dependent antioxidant enzyme critical for neutralizing lipid peroxides, thereby preventing ferroptosis, may be involved in ferroptosis in non-obstructive azoospermia (NOA) [14]. In semen samples from asthenozoospermic patients, the levels of GPX4 and solute carrier family 7 member 11 (SLC7A11), which is a cystine/glutamate antiporter essential for glutathione synthesis, acting as a gatekeeper of ferroptosis susceptibility, were found to be lower than those in normozoospermic individuals, while reactive oxygen species (ROS), malondialdehyde (MDA), and iron concentrations were significantly elevated. A strong positive correlation between Gpx4 mRNA levels and sperm motility was observed, suggesting that reduced GPX4 expression may be a key factor in the ferroptosis mechanism underlying male infertility [15]. Varicocele (VC), a prevalent cause of male infertility, is characterized by abnormal dilation of the pampiniform venous plexus, leading to impaired thermoregulation, oxidative stress (e.g., reduced glutathione peroxidase activity), and compromised spermatogenesis [16,17]. Furthermore, in diabetic patients, sperm motility is decreased, while ROS and lipid peroxidation are increased. Both type 1 and type 2 diabetes mellitus lead to low ejaculation volume and decreased sperm motility [18]. Additionally, hepatitis infections can cause sperm defects, and exposure to hepatitis B surface antigens can downregulate GPX4 expression in Sertoli cells, indicating that hepatitis may induce male infertility via ferroptosis in Sertoli cells [19].
Ferroptosis also plays a role in male infertility induced by environmental pollutants (Figure 2). Studies indicate that exposure to particulate matter(PM) such as PM2.5 and PM10, cigarette smoke, cadmium (Cd), arsenite, and other environmental stressors [20,21,22,23,24,25,26,27,28,29,30,31,32] triggers ferroptosis in testicular cells, especially in Sertoli cells [23,28,33], Leydig cells [22], and germ cells [20,21,24]. Heavy metals, such as Cd, significantly impact nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 has been identified as a master regulator of antioxidant defense, activating GPX4 to counteract oxidative stress. Beyond ferroptosis, Nrf2 dysregulation is implicated in neurodegenerative diseases, cancer, and metabolic disorders, underscoring its broad pathophysiological relevance [34]. Cd exposure increases Nrf2 expression and activates its downstream signaling pathways. By contrast, zinc (Zn) counteracts Cd-induced damage by increasing metabolically active Sertoli cells and alleviating Sertoli cell injury, partially through Nrf2 regulation [35]. Hydrogen sulfide also prevents ferroptosis in Sertoli cells by suppressing intracellular acrolein accumulation [33]. This is associated with ROS accumulation, disruption of iron homeostasis, and subsequent activation of ferroptosis. The molecular mechanisms involve the upregulation of ferroptosis-related genes and proteins, such as ferredoxin 1 (FDX1), which has been identified as a mitochondrial iron-sulfur protein implicated in cuproptosis, linking copper toxicity to lipoylated tricarboxylic acid (TCA) cycle enzyme aggregation [36] and the downregulation of antioxidant defenses, leading to lipid peroxidation and cell death [22,27,28,29,37]. Antioxidants or ferroptosis inhibitors have shown potential in mitigating testicular toxicity and improving sperm quality, making ferroptosis inhibition a promising strategy for treating male infertility caused by environmental exposure [21,24,33]. Related randomised controlled trials suggest antioxidant supplementation may improve live birth rates in subfertile males [38].
Furthermore, ferroptosis inhibitors or natural extracts may improve male infertility by suppressing ferroptosis. Ferroptosis inhibitors mainly exert their effects by inhibiting iron accumulation and lipid peroxidation [39]. Ferrostatin-1 (Fer-1) and deferoxamine (DFO), two well-characterized ferroptosis inhibitors, mitigate oxidative stress and lipid peroxidation in testicular cells [40,41]. In busulfan-induced oligospermia models, Fer-1 restored sperm quality by upregulating Gpx4 and modulating Nrf2 signaling, underscoring its therapeutic potential in male infertility [40,41]. Similarly, compounds such as ursonic acid [42], aucubin [43], cordycepin [44], myristic acid [45], hirsutella sinensis [46], mangiferi [47], and Semen Cuscutae and Fructus Lycii [48] have demonstrated protective effects against spermatogenic damage by modulating ferroptosis and enhancing antioxidant defenses, highlighting their potential therapeutic value in treating male infertility. These findings underscore the importance of targeting ferroptosis as a therapeutic approach for male infertility.

2.2. Mechanisms of Ferroptosis in Male Infertility

Key factors in the ferroptosis pathway mediate male infertility through the following mechanisms. ROS, byproducts of aerobic metabolism, include superoxide anions (O2), hydroxyl radicals (•OH), hydrogen peroxide (H2O2), and singlet oxygen (1O2). In the presence of mitochondrial superoxide dismutase (SOD, EC 1.15.1.1), O2 is converted to H2O2, which then diffuses from mitochondria into the cytoplasm. At high iron concentrations that favor the Fenton reaction, H2O2 generates highly reactive O2 groups, and further peroxide accumulation induces ferroptosis in germ cells [49]. Imbalances in ROS production and antioxidant defenses are key contributors to male infertility. ROS and calcium (Ca2+) homeostasis disturbances activate mitochondrial permeability transition pores (mPTP), leading to mitochondrial dysfunction in sperm [50]. Studies in infertility models have shown that elevated peroxides levels and excessive ROS accumulation promote lipid peroxidation and subsequent ferroptosis [22,24,29,51]. Collectively, ROS accumulation is a major driver of ferroptosis-mediated male infertility.
Glutathione (GSH) is a tripeptide thiol compound synthesized via cysteine transport into cells through the system Xc glutamate antiporter. GSH exerts antioxidant effects by scavenging lipid peroxides, reducing ROS production, and inhibiting ferroptosis [52]. Glutathione synthase (GSS, EC 6.3.2.3) catalyzes the final step in GSH synthesis. In GSS-deficient cells, compromised antioxidant capacity leads to lipid peroxidation and ferroptosis. This deficiency downregulates GPX4 and upregulates arachidonic acid 15 lipoxygenase (ALOX15), a downstream target suppressed by GPX4. These changes trigger irreversible plasma membrane rupture, exacerbating ferroptosis and contributing to male infertility [53]. In S8/Gss−/− mice, decreased Gpx4 and increased Alox15 levels caused testicular ferroptosis, abnormal spermatogenesis, reduced sperm concentration, and morphological defects, ultimately leading to infertility [54]. Increasing the GSH level mitigated these effects by suppressing ROS production, inhibiting ferroptosis, and restoring spermatogenesis [55]. GPX4 reduces phospholipid hydroperoxides (PLOOHs) to their corresponding phospholipid alcohols (PLOHs), thereby inhibiting ferroptosis. GPX4 downregulation promotes germ cell ferroptosis, adversely affecting male fertility [49,56]. In NOA patients, GPX4 downregulation is associated with ferroptosis [14]. Similarly, asthenozoospermic patients exhibit GPX4 and SLC7A11 downregulation, which correlates with increased iron deposition and impaired sperm motility [15]. Enhancing Gpx4 expression alleviates ferroptosis-related infertility [51]. The ferroptosis inhibitor Fer-1 significantly reduced lipid ROS and restored Gpx4 levels in GC-2spd cells [21]. Additionally, fecal microbiota transplantation upregulated Gpx4, inhibited ferroptosis, and restored spermatogenesis in mice [57].
Critical factors, including transferrin (TF) and Nrf2, mediate ferroptosis in testicular cell. Cells regulate iron intake through TF and transferrin receptor (TFRC). Experimental studies have shown that knockdown of Tfrc can block iron-dependent cell death by reducing mitochondrial and intracellular Fe2+ levels in TM4 cells, suggesting that iron overload plays a critical role in ferroptosis in these cells [23]. Additionally, Nrf2 is a key regulator of ferroptosis, directly or indirectly upregulating GPX4 expression and enhancing ferroptosis resistance. Nrf2 promotes intracellular cysteine enrichment via system Xc, enhances GPX4 synthesis, and reduces its degradation [53]. Cancer cells with elevated NRF2 levels exhibit increased ferroptosis resistance [58]. NRF2 is essential for maintaining iron homeostasis and inhibiting ferroptosis through GPX4 upregulation [58]. The absence of Nrf2 downregulates GPX4, impairing the clearance of phospholipid hydroperoxides (PLOOHs) and causing excessive lipid peroxide accumulation, thereby triggering ferroptosis in spermatogenic cells [59]. Functional polymorphisms in NRF2 promoters are strongly associated with defective spermatogenesis in humans [60,61]. And sperm from oligozoospermic men show lower NRF2 and GPX4 expression compared to healthy controls, further supporting this mechanism [62,63]. Ferroptosis represents a pivotal pathway in iron-induced male infertility, integrating the accumulation of lipid ROS and lipid peroxidation, mitochondrial dysfunction, and genetic dysregulation.

3. Cuproptosis

3.1. Cuproptosis in Male Infertility

Copper (Cu) is an essential trace element crucial for male reproductive health, particularly in spermatogenesis. Elevated Cu levels in seminal plasma have been associated with impaired sperm motility and abnormal morphology, likely due to its involvement in oxidative stress. Subfertile men exhibit significantly higher Cu concentrations in their serum, seminal plasma, and urine compared to fertile controls, which correlate with lower sperm count and motility [64,65]. Furthermore, Yin et al. [66] demonstrated that high Cu levels in seminal plasma were associated with increased oxidative stress markers, such as MDA, and a higher risk of idiopathic oligoasthenoteratozoospermia. The Cu/Zn ratio in seminal plasma also appears to be a crucial factor in male infertility. Akinloye et al. [67] noted that infertile men exhibit a lower Cu/Zn ratio in seminal plasma, suggesting that an imbalance between these trace elements could impair sperm function. Collectively, these studies highlight the importance of regulating Cu levels to maintain sperm health and fertility. Therefore, determination of Cu levels in serum, seminal plasma, and urine during infertility investigations is recommended. The toxicity of Cu is also related to the destruction of enzymes containing iron-sulfur (Fe-S) clusters. Multiple studies have shown that Cu blocks the formation of Fe-S clusters and disrupts the stability of Fe-S proteins. The accumulation of Cu ions can trigger oxidative stress. Excessive Cu leads to impaired sperm production and infertility through ROS generation and loss of mitochondrial membrane potential [68].

3.2. Possible Mechanisms of Cuproptosis in Male Infertility

Cuproptosis, a Cu-dependent form of programmed cell death, has emerged as a critical mechanism underlying male infertility, particularly under conditions of Cu overload. This process is characterized by mitochondrial dysfunction, oxidative stress, and aggregation of lipoylated proteins, leading to irreversible germ cell damage. Studies show that Cu nanoparticles (CuNPs) and copper sulfate (CuSO₄) induce oxidative stress, leading to increased MDA levels and apoptosis in testicular cells. These effects reduce sperm quality, disrupt hormone levels, and impair testicular function, with higher doses resulting in more severe damage [69,70,71]. CuNPs, for instance, downregulate steroid receptors, which inhibits germ cell proliferation and compromises spermatogenesis [70,72]. In a murine model of Cu overload (CuSO4 exposure), Zhang et al. [73] demonstrated that elevated Cu levels disrupted testicular Cu homeostasis, upregulated cuproptosis-related factors (e.g., Fdx1), and induced mitochondrial ATP depletion, resulting in apoptosis of spermatogenic cells. Similarly, Guo et al. [74] observed that Cu-induced oxidative stress triggered DNA damage and suppressed DNA repair pathways, further exacerbating germ cell apoptosis. Mitochondrial damage, evidenced by reduced membrane potential and increased ROS production, was also linked to autophagy activation in pig testes exposed to long-term Cu exposure [75]. These findings highlight the centrality of mitochondrial dysfunction in cuproptosis-mediated spermatogenic failure, with oxidative stress acting as a key amplifier of cellular damage.
Beyond direct cellular damage, Cu overload perturbs immune homeostasis and epigenetic regulation in the testicular microenvironment. Zhao et al. [76] identified upregulated expression of cuproptosis-related genes (e.g., NLR family pyrin domain-containing 3 (NLRP3), solute carrier family 31 member 1 (SLC31A1)) in human spermatogenic dysfunction, correlating with immune cell infiltration, particularly resting memory CD4+ T cells. This inflammatory response, driven by toll like receptor 4(TLR4) /NF-κB signaling, synergizes with PANoptosis (pyroptosis, apoptosis, and necroptosis) to exacerbate testicular damage [77]. Epigenetic modifications further compound these effects. Shaoyong et al. [78] reported that Cu oxide nanoparticles reduced H3K9me3 levels in spermatozoa, impairing mitochondrial energy metabolism and transgenerationally transmitting asthenospermia. Additionally, Huang et al. [79] linked Cu overload to lncRNA:CR43306 deficiency in Drosophila, which disrupted spermatid differentiation via ferroptosis-cuproptosis crosstalk. These studies underscore the interplay between Cu toxicity, immune dysregulation, and epigenetic instability in male infertility, suggesting that therapeutic strategies must address both direct and systemic mechanisms.
Current research highlights potential interventions to mitigate Cu-induced spermatogenic damage. Chen et al. [80] demonstrated that GSH supplementation alleviated oxidative stress and apoptosis in Wilson’s disease models by restoring mitochondrial function. Similarly, retinoic acid rescued meiosis initiation in NSC319726-exposed mice, independent of cuproptosis, by compensating for retinol dehydrogenase deficiency [81]. Natural compounds like propolis and Tribulus terrestris extract attenuated Cu toxicity by enhancing antioxidant defenses and steroidogenesis [82,83]. Cu chelators (e.g., penicillamine, DMSA) and TLR4 inhibitors (e.g., eritoran) also showed efficacy in reducing testicular Cu deposition and inflammation [77,84,85]. However, challenges persist in managing chronic exposure and transgenerational effects. For instance, Nicy et al. [72] observed persistent testicular impairment in mice even after cessation of Cu nanoparticle treatment, indicating irreversible damage at high doses.
Cuproptosis represents a pivotal pathway in Cu-induced male infertility, integrating mitochondrial dysfunction, oxidative stress, immune activation, and epigenetic dysregulation. Copper homeostasis has been proven to regulate various cellular activities in cancer, and Cu chelators have huge potential for treatment [86]. Clinical trials on bis-choline tetrathiomolybdate (TTM), a copper chelator, demonstrate reduced copper uptake in Wilson’s disease and tolerability in breast cancer patients [87,88]. While therapeutic advances such as antioxidants and chelators show promise, gaps remain in understanding tissue-specific Cu transporters (e.g., high-affinity Cu transporter CTR1(CTR1), ATPase copper transporting alpha(ATP7A)) and their roles in germ cell survival [89,90,91]. Bioinformatic analyses revealed that elevated FDX1 expression correlates with poor prognosis in glioma and gastric cancer, suggesting its role in copper-driven cell death [92,93]. However, the clinical relevance of cuproptosis-related biomarkers (e.g., NRF2) in human nonobstructive azoospermia warrants validation in larger cohorts [94]. Multidisciplinary approaches combining omics technologies, targeted drug delivery, and epigenetic interventions will be essential to translate these findings into effective therapies for Cu-related reproductive disorders.

4. Autophagy

4.1. Autophagy Exists in Clinical Diseases Related to Male Infertility

Autophagy is a conserved lysosome-dependent process wherein cytoplasmic components (e.g., damaged organelles) are sequestered within double-membraned autophagosomes and degraded to maintain cellular homeostasis. In autophagy, multiple key factors work together to ensure autophagosome formation, expansion, and fusion with lysosomes. Beclin-1(BECN1) initiates autophagosome formation by interacting with the phosphatidylinositol 3-kinase complex. Sequestome 1 (sequestome 1, also known as p62) recruits proteins to participate in the formation of autophagosomes.Autophagy-related 7 (ATG7), an E1-like activating enzyme, promotes autophagosome membrane expansion by facilitating autophagy-related 5 (ATG5) and ATG12 (autophagy-related protein 12) binding. ATG5 and ATG12 form a complex that binds with autophagy-related 16-like 1 (ATG16L1), further extending and closing the autophagosome membrane. Microtubule-associated protein 1 light chain 3 (LC3 proteins), markers of autophagosome membranes, convert from soluble LC3-I to membrane-bound LC3-II, integrating LC3-II into autophagosome membranes to aid their formation and expansion. Lysosome-associated membrane protein 2 (LAMP2), a crucial lysosomal membrane protein, is involved in the fusion of autophagosomes with lysosomes. GABA type A receptor-associated protein (GABARAP) is a ubiquitin-like protein belonging to the ATG8 family, involved in autophagosome formation, expansion, and fusion with lysosomes [95,96,97]. PTEN-induced putative kinase 1 (PINK1), a serine/threonine kinase, is a sensor of mitochondrial damage and mitochondrial autophagy (mitophagy). These factors collaborate in autophagy to maintain cell homeostasis.
Autophagy has been implicated in clinical diseases associated with male infertility. The CT55 gene of two infertile siblings had a semizygous nonsense mutation, with symptoms of insufficient sperm personalization, excessive cytoplasmic residue, and defective acrosome development. Ct55-knockout male mice also showed infertility and sperm destruction. Endoplasmic reticulum (ER)- and ribosome-associated proteins were increased in the testes of patients and mice, while both LAMP2 and GABARAP were significantly reduced, indicating decreased autophagy activity. Further research showed that CT55 participates in autophagy through its interaction with LAMP2 and GABARAP, thereby affecting spermatogenesis [98]. In addition, patients with NOA have lower levels of MIR-188-3p and higher levels of ATG7 in testicular tissue. A further study revealed that the inhibition of MIR-188-3p directly targeted ATG7 to increase its expression and promote autophagy, impairing spermatogenesis [99]. In a study of infertility mediated by varicocele, the levels of autophagy markers ATG7 and LC3 proteins in the semen of patients were significantly higher than in the semen of fertile men, suggesting that autophagy is overactivated in male infertility caused by varicocele [100]. In the testicular tissue of rats with varicocele, the levels of autophagy-related protein LC3 and LC3-II/LC3-I ratio were significantly increased, further confirming this observation [101]. In conclusion, autophagy levels are abnormal in the clinical disease of male infertility mentioned above, which affects spermatogenesis.
Environmental pollutants disrupt spermatogenesis by dysregulating autophagy through oxidative stress, endoplasmic reticulum (ER) stress, and alterations in signaling pathways. Mycotoxins, such as zearalenone (ZEA) and aflatoxin B1 (AFB1), inhibit the PI3K/Akt/mTOR axis, activating autophagy in Sertoli and germ cells. ZEA induced oxidative stress in goat Sertoli cells, elevating the LC3-II/LC3-I ratio, reducing p62, and promoting lysosomal biogenesis via transcription factor EB (TFEB) nuclear translocation [102]. Similarly, AFB1 increased autophagosome formation and upregulated LC3, Beclin-1, and Atg5 in mouse testes by suppressing PI3K/Akt/mTOR, with oxidative stress further exacerbating autophagy [103]. Heavy metals like Cd and Cu exhibit dual roles: Cd hyperactivates autophagy by elevating LC3, Atg5, and Lamp2, disrupting the blood–testis barrier (BTB) through vimentin degradation [104,105], while Cu activates AMPK-mTOR-dependent autophagy to mitigate oxidative damage, paradoxically protecting sperm motility [106]. Nanoparticles and endocrine disruptors further modulate autophagy. Zinc oxide nanoparticles (ZnO NPs) induce ROS-mediated autophagy in GC-1 spermatogonia, increasing LC3-II and Atg5 [107]. Di-(2-ethylhexyl) phthalate (DEHP) upregulates PINK1/PARKIN and XBP1, driving mitophagy in spermatogonia via ER stress [108], whereas tributyltin chloride (TBTCL) inhibits autophagy, triggering apoptosis in Sertoli cells [109]. Perfluorooctanoic acid (PFOA) blocks autophagosome-lysosome fusion by downregulating α-SNAP, impairing BTB integrity [110]. Ethanol selectively activates autophagy in stage VII–VIII Sertoli cells, marked by LC3 accumulation and PINK1-mediated mitochondrial clearance, disrupting spermatogenic cycles [111]. These findings underscore a unifying mechanism: pollutants induce oxidative/ER stress to dysregulate autophagy, either promoting excessive degradation (e.g., Cd, ZEA) or impairing flux (e.g., PFOA), ultimately compromising germ cell viability and BTB function. Targeting autophagy regulators such as mTOR or TFEB may offer therapeutic strategies to counteract pollutant-induced male infertility.
Autophagy plays a crucial role in treating male infertility and spermatogenic disorders induced by lifestyle factors (e.g., obesity), drugs, or environmental factors. For example, Qiangjing tablets activated mitophagy in a mouse model of asthenozoospermia, improving sperm motility by enhancing the LKB1/AMPK/ULK1 signaling pathway [112]. Similarly, eprosartan activated autophagy in testicular torsion models by modulating the Beclin-1 and AMPK/mTOR pathways, mitigating testicular damage [113]. Clinical study have found that infertile oligoasthenoteratozoospermic men associated with varicocele have significantly higher cystatin C levels in semen than healthy males. And six months after surgery, the seminal cystatin C levels were significantly lower than pre-operative levels.This implied that cystatin C may ameliorate male infertility due to varicocele by modulating autophagy [114]. Sancai Lianmei granules improve sperm density and viability in obese mice by inhibiting excessive autophagy [115]. In drug-induced infertility, such as busulfan or paclitaxel exposure, autophagic activity is dysregulated, and supplementation with fatty acids like octanoic acid or compounds like naringin can reverse these effects, improving spermatogenesis and testicular function [116,117]. Moreover, thymoquinone alleviates cisplatin-induced testicular damage by modulating autophagic and apoptotic pathways, restoring sperm quality [118]. Environmental pollutants like bisphenol A and Cd also disrupt spermatogenesis via autophagy dysregulation. Lactoferrin and MSC-derived exosomes help to restore autophagic flux, reducing oxidative stress and improving testicular function [119,120]. Berberine nanostructured biloalbuminosomes and M1 nanostructured biloalbuminosomes ameliorated Cd- and Pb-induced testicular and prostate damage in rats by attenuating oxidative stress-induced excessive autophagy [121]. Ginkgo biloba extract attenuated insecticide deltamethrin-induced testicular damage in mice by upregulating SKP2, a key regulator of Beclin-1-independent autophagy, and inhibited Beclin-1 activation to regulate autophagy [122]. These studies highlight the potential of autophagy modulation as a therapeutic strategy for male infertility caused by various factors.

4.2. Mechanism of Autophagy in Male Infertility

Autophagy plays a pivotal role in meiosis and early spermatogenesis, ensuring genomic integrity and germ cell survival. Forkhead box J2 (Foxj2), a transcription factor critical for meiotic progression, regulates DNA double-strand break (DSB) repair and chromosomal synapsis. Germline deletion of Foxj2 in mice resulted in meiotic arrest due to defective DSB repair and reduced expression of repair factors (e.g., Rad51, Brca1), highlighting its role in maintaining meiotic fidelity [123]. Paradoxically, Foxj2 overexpression activated aberrant autophagy by upregulating Lamp2A, a chaperone-mediated autophagy receptor, leading to excessive lysosomal degradation and meiotic failure [124]. Concurrently, adenine nucleotide translocase 4 (Ant4), essential for mitochondrial ADP/ATP transport, is indispensable for spermatocyte survival. Ant4-deficient mice exhibited spermatocyte apoptosis, mitochondrial dysfunction, and disrupted autophagic flux via the AKT-AMPK-mTOR pathway, linking energy metabolism to autophagy regulation [125,126]. These findings underscore the delicate balance of autophagy in meiosis: while basal autophagy supports DSB repair and mitochondrial homeostasis, dysregulation—either deficiency or hyperactivation—compromises germ cell viability.
During spermiogenesis, autophagy facilitates cytoplasmic remodeling and organelle clearance, processes critical for sperm individualization. ATG5, a core autophagy protein, mediates autophagosome formation and sperm elongation. Conditional knockout of Atg5 in germ cells disrupted spermatid development, resulting in residual cytoplasmic bodies, malformed acrosomes, and infertility [127]. Similarly, BECN1, a key regulator of autophagosome nucleation, is essential for spermiation and mitochondrial organization. Germline Becn1 deletion impaired autophagy, leading to abnormal sperm motility, acrosome defects, and epididymal epithelial loss [128]. Beyond canonical autophagy, Atg5 also participates in non-autophagic processes such as lysosomal repair via ESCRT machinery, which is critical for maintaining sperm structural integrity [129]. Additionally, cathepsin B (CTSB), a lysosomal protease, modulates autophagy–apoptosis crosstalk during sperm maturation. Ctsb-null mice exhibited inhibited autophagy, increased apoptosis, and epididymal epithelial damage, emphasizing CTSB’s role in balancing degradation and survival pathways [130,131]. mTOR acts as a core factor regulating spermatogenesis, and the use of mTOR inhibitors such as rapamycin can improve spermatogenesis by activating autophagy in the presence of autophagy deficiency [132,133]. These studies collectively demonstrate that autophagy coordinates organelle clearance, energy homeostasis, and stress responses to ensure functional sperm production.
Autophagy in spermatogenesis is tightly regulated by phosphorylation events and metabolic signaling. BECN1 phosphorylation fine-tunes autophagosome assembly, integrating stress signals to maintain pro-survival autophagy [134]. For instance, oxidative stress and ER stress (e.g., induced by environmental toxins) modulate BECN1 activity, impacting spermatogonial survival and meiotic progression. However, unresolved questions persist: (1) How do Foxj2 and Ant4 mechanistically intersect with autophagy pathways? (2) What role do non-canonical autophagy proteins, such as ATG5 in lysosomal repair, play in sperm quality? (3) Can CTSB or LAMP2A be targeted and lysosomal stabilizers rescue autophagy dysregulation in infertility by mitigating environmental or genetic insults? Understanding these mechanisms will advance strategies to address male infertility linked to autophagy dysfunction.

5. Pyroptosis

5.1. Pathological Roles of Pyroptosis in Diverse Etiologies of Male Infertility

Pyroptosis, a pro-inflammatory programmed cell death driven by gasdermin family proteins (e.g., gasdermin D (GSDMD)), is characterized by plasma membrane pore formation, cytokine release (e.g., interleukin-1β(IL-1β)), and osmotic lysis. It plays dual roles in testicular immune defense and pathological inflammation. Clinical and preclinical studies implicate pyroptosis as a hallmark of spermatogenic failure. In human spermatogenic dysfunction, pro-pyroptotic factor CASPASE-4(CASP4) and anti-pyroptotic factor GPX4 are identified as key regulators, with CASP4 negatively correlating with Johnsen scores and GPX4 exerting protective effects [135]. Varicocele-associated infertility involves oxidative mtDNA activating the cGAS/STING pathway, which promotes NLR family pyrin domain-containing 3 inflammasome (NLRP3 inflammasome) activity and pyroptosis in Sertoli cells [136]. Seminal inflammasome markers (e.g., IL-1β, cysteine-aspartic acid protease-1(caspase-1)) are elevated in varicocele patients, and varicocelectomy reduces these mediators, thereby improving semen quality [137]. Obesity and metabolic disorders exacerbate pyroptosis through NLRP3 inflammasome activation. A high-fat diet (HFD) elevated testicular high mobility group box 1 (HMGB1) and NLRP3 levels, impairing sperm parameters, whereas zinc supplementation mitigated oxidative stress and inflammation [138]. Blautia wexlerae depletion in obesity disrupts acetate-mediated suppression of NLRP3, whereas its restoration attenuates pyroptosis and improves fertility [139]. These data underscore pyroptosis as a pathological nexus in male infertility, driven by oxidative stress, cytokine dysregulation, and environmental insults.
The environmental drivers of testicular pyroptosis are various. Environmental toxins such as fluoride and 1,2-dichloroethane (1,2-DCE) induce pyroptosis via IL-17A signaling and piRNA-mmu-1019957/IRF7 pathways, respectively, thereby impairing germ cell viability [140,141]. Co-exposure to microcystin and nitrite synergistically enhances mitochondrial ROS (mtROS)-dependent pyroptosis and apoptosis, exacerbating spermatogenic disorders [142]. Silver nanoparticles (AgNPs) induce NF-κB/NLRP3-driven inflammation in spermatogonia, highlighting their reprotoxic potential [143]. Additionally, lead exposure triggers pyroptosis-mediated fibrosis via NF-κB and caspase-1 activation, further compromising testicular function [144]. These studies emphasize the interplay between extrinsic factors and intrinsic inflammasome pathways in driving male reproductive dysfunction. The different forms of cell death implicated in environmental pollutant-induced male infertility are summarized in Table 1.

5.2. Molecular Mechanisms of Pyroptosis and Its Role in Testicular Homeostasis

Under physiological conditions, pyroptosis aids in eliminating infected or damaged cells, maintaining immune homeostasis [147]. However, pathological activation of the NLRP3 inflammasome, triggered by oxidative stress or mitochondrial dysfunction, disrupts spermatogenesis by cleaving caspase-1/4/11, which processes gasdermin D (GSDMD) to form membrane pores, releasing pro-inflammatory cytokines (IL-1β, IL-18) and cellular debris [148,149]. For instance, in Sertoli cell-only syndrome (SCOS), upregulated CASP1 and CASP4 in Leydig and Sertoli cells drive pyroptosis, which correlates with germ cell loss and elevated inflammatory markers [150]. Similarly, Cd exposure induces absent in melanoma 2 inflammasome (AIM2)-dependent pyroptosis via m6A-modified Lonp1, which disrupts mitochondrial proteostasis and releases mitochondrial DNA (mtDNA) into the cytoplasm, exacerbating testicular injury [145]. These findings highlight the NLRP3/GSDMD axis as central to pyroptosis-mediated testicular damage, with caspase activation and mitochondrial dysfunction as critical contributors. Pyroptosis emerges as a pivotal mechanism linking inflammation, oxidative stress, and environmental toxins to male infertility. Moreover, context-dependent regulators like miRNAs (e.g., MiR-153, MiR-195-5p) and mitochondrial dynamics modulate outcomes [151,152].
Targeting pyroptosis pathways offers promising interventions. Melatonin and Bifidobacterium suppress NLRP3 and caspase-1/11, alleviating fluoride- and 1,2-DCE-induced damage [140,141]. Chlorogenic acid inhibits cGAS/STING-mediated NLRP3 activation in varicocele, restoring mitochondrial homeostasis [136]. Pharmacological GSDMD inhibitors (e.g., disulfiram) and gut microbiota modulators like Guijiajiao improve spermatogenesis by reducing inflammation [153,154] (Table 2). Monosodium glutamate upregulates pyroptotic markers (NLRP3, caspase-3), which resveratrol reverses via antioxidant and anti-pyroptotic effects [146]. NLRP3 inhibitors (e.g., MCC950) attenuate COVID-19-associated testicular inflammation [155]. Virtual screening identified FDA-approved drugs (ChEMBL IDs: 4297185, 1201749, 1200545) targeting NLRP3 for varicocele treatment [156]. Therapeutic strategies targeting pyroptosis components show preclinical efficacy, yet clinical validation is imperative. Future research should prioritize cell-specific delivery systems for pyroptosis inhibitors and combinatorial therapies targeting oxidative stress and mitochondrial integrity.
However, challenges remain: the tissue-specific roles of gasdermin isoforms (e.g., gasdermin E ) in testicular pyroptosis are underexplored, and crosstalk between pyroptosis, apoptosis, and autophagy in germ cells requires further study [157,158]. Understanding pyroptosis’s dual roles—pathological vs. protective—in testicular physiology will refine interventions, ultimately improving fertility outcomes for affected individuals.

6. The Relationship Between Cell Death Forms in Male Infertility

6.1. The Dual Role of SLC7A11 in Ferroptosis and Disulfidptosis

Disulfidptosis is a novel form of cell death. In recent years, with the in-depth study of cell death mechanisms, the role of disulfidptosis in male infertility has gradually been revealed. High expression of SLC7A11 (responsible for cysteine uptake) promotes the depletion of nicotinamide adenine dinucleotide phosphate (NADPH). Under glucose starvation, SLC7A11-mediated cysteine uptake further depletes intracellular NADPH, leading to its depletion and the accumulation of disulfide bonds. This results in the formation of disulfide bonds between actin cytoskeleton proteins and the breakdown of the actin filament (F-actin) network [159,160], which leads to disulfidptosis.
SLC7A11, a critical component of the cystine/glutamate antiporter system Xc, plays a paradoxical role in regulating ferroptosis and disulfidptosis, two distinct forms of regulated cell death implicated in male infertility. Ferroptosis, driven by iron-dependent lipid peroxidation, is tightly linked to SLC7A11’s function in maintaining redox homeostasis. By importing cystine for GSH synthesis, SLC7A11 supports GPX4 activity to neutralize lipid peroxides, thereby protecting testicular cells from oxidative damage [33,161,162,163]. For instance, Cr (VI) and DEHP exposures downregulate SLC7A11, deplete GSH, and induce ferroptosis in Sertoli and Leydig cells, leading to seminiferous tubule atrophy and impaired spermatogenesis [161,162,164]. Conversely, disulfidptosis arises from excessive cystine accumulation under glucose deprivation, where SLC7A11 overexpression overwhelms NADPH-dependent reduction capacity, causing toxic disulfide stress and membrane rupture [165,166]. In Bisphenol F(BPF)-induced testicular injury, obesity-associated protein(FTO) deficiency disrupts m6A-mediated SLC7A11 mRNA stability, exacerbating disulfidptosis via YTH domain family 2(YTHDF2)-dependent decay [165]. These findings highlight SLC7A11’s dual regulatory role: its downregulation promotes ferroptosis by limiting antioxidant defenses, while its overexpression under metabolic stress triggers disulfidptosis through cysteine overload.

6.2. The Facilitating Role of ROS in Ferroptosis and Cuproptosis

Common mechanisms in both ferroptosis and cuproptosis are oxidative stress and mitochondrial dysfunction. Both ferroptosis and cuproptosis trigger cell damage through ROS accumulation. In ferroptosis, Fe2+ generates hydroxyl radicals (OH•) through the Fenton reaction, resulting in lipid peroxidation [49,52]. In cuproptosis, excess Cu2+ directly produces ROS that disrupt mitochondrial membrane potential [68,73]. GSH depletion and downregulation of GPX4 in ferroptosis [15,54] and GSH supplementation in cuproptosis [80] suggest that both are dependent on the integrity of the antioxidant system. Ferroptosis leads to mitochondrial lipid peroxidation through GPX4 inhibition failure [53], whereas cuproptosis induces mitochondrial ATP depletion and loss of membrane potential via Cu ions [73]. Both involve the activation of mitochondrial permeability transition pores (mPTP), which triggers cell death [50,75].
Through cross-regulation at the molecular level, Nrf2 inhibits lipid peroxidation by upregulating GPX4 and SLC7A11 in ferroptosis [53,58], and in cuproptosis, activation of Nrf2 alleviates Cu-induced oxidative stress [80]. Whereas FDX1 regulates lipid peroxidation in ferroptosis [22] and is a key factor in Cu-dependent death in cuproptosis [73]. Translating these findings to male infertility, we hypothesize that FDX1 overexpression in testicular cells may exacerbate copper toxicity, impairing spermatogenesis. Future studies should explore urinary/serum copper levels and FDX1 activity in infertile males, potentially establishing FDX1 as a diagnostic biomarker or therapeutic target. Cu overload triggers cuproptosis through the mitochondrial TCA cycle, facilitating intracellular interactions with ferroptosis, thereby governing testicular aging [79]. Ferroptosis and cuproptosis cross-function in male infertility through oxidative stress, mitochondrial damage, and Nrf2 pathway dysregulation. In the future, it is necessary to further explore the interaction mechanism of FDX1 and GPX4 molecules in the two types of death to develop precise intervention methods.

6.3. The Balance of Pyroptosis and Autophagy in Male Fertility

The balance between pyroptosis and autophagy is essential for male fertility. Autophagy inhibits pyroptosis hyperactivation by degrading the NLRP3 inflammasome, e.g., chlorogenic acid inhibits NLRP3 through the cGAS/STING pathway and ameliorates varicocele [136]. Conversely, IL-1β released through pyroptosis may inhibit autophagy, creating a vicious cycle [137]. Pyroptosis and autophagy share the upstream signaling pathways mTOR and ROS. mTOR inhibits the activation of autophagy (e.g., the AMPK/mTOR axis regulates autophagy) while inhibiting NLRP3 inflammasome activity [104,106]. ROS activates the NLRP3 inflammasome to trigger pyroptosis (e.g., mtROS accumulation in varicocele [136]), while inducing autophagy to clear the damaged component. If autophagy is insufficient (e.g., antioxidant deficiency), pyroptosis predominates and spermatogenic cell death (e.g., microcystins synergize with nitrite) [142]. Crossover mechanisms occur at the molecular level. Cleavage fragments of GSDMD may interfere with autophagosome maturation by disrupting lysosomal membranes [149]. Synergy and conflict of blood–testis barrier function are induced by pyroptosis and autophagy. Sertoli cell pyroptosis directly disrupts the blood–testis barrier, leading to immune cell infiltration (e.g., lead exposure activates pyroptosis via NF-κB/caspase-1) [144]. Autophagy maintains cellular junctions by degrading aberrant proteins such as connexin 43. For example, perfluorooctanoic acid (PFOA) blocks autophagosome–lysosome fusion, impairing the blood–testis barrier [110].

7. Conclusions

This review delineates the pathogenic roles of ferroptosis, cuproptosis, autophagy, and pyroptosis in male infertility, highlighting their molecular cross-talk and environmental triggers. Key mechanisms include oxidative stress, mitochondrial dysfunction, and inflammasome activation, which disrupt spermatogenesis and BTB integrity (Figure 3). Therapeutic strategies targeting antioxidant defenses (e.g., Nrf2 activators), metal homeostasis, and inflammatory pathways demonstrate efficacy in mitigating testicular damage.
Future research must prioritize: (1) validating biomarkers (e.g., GPX4, FDX1) in clinical cohorts for early diagnosis; (2) developing precision therapies, such as nanoparticle-based delivery of pathway-specific inhibitors (e.g., GSDMD or FDX1-targeted drugs) or CRISPR editing to correct autophagy-related mutations; (3) elucidating tissue-specific regulators (e.g., gasdermin E in pyroptosis) and their interactions; (4) addressing the transgenerational effects of environmental toxins through epigenetic studies (e.g., H3K9me3 loss in CuONP exposure); (5) multi-omics integration to map the multi-cell death pathway dynamics across spermatogenic stages. Integrating multi-omics and AI-driven models will advance mechanistic understanding and therapeutic innovation, potentially improving fertility outcomes in affected populations.

Author Contributions

D.C., J.L., Z.P., R.F., C.C., F.L. and C.L. wrote the manuscript. Y.L. and Y.S. were responsible for revising the manuscript. W.C. was responsible for study conception, revising the manuscript, and final approval of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was fundede by grants from the Natural Science Foundation of Hunan Province (2023JJ30424, 2024JJ5284), Scientific Research Project of the Hunan Provincial Department of Education (24C0023), and Hunan Province College Students Research Learning and Innovative Experiment Project (S202310542141).

Data Availability Statement

The data are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no competing financial interests.

References

  1. WHO TEAM. Sexual and Reproductive Health and Research. Infertility Prevalence Estimates, 1990–2021; WHO: Geneva, Switzerland, 2023. [Google Scholar]
  2. Levine, H.; Jørgensen, N.; Martino-Andrade, A.; Mendiola, J.; Weksler-Derri, D.; Jolles, M.; Pinotti, R.; Swan, S.H. Temporal trends in sperm count: A systematic review and meta-regression analysis of samples collected globally in the 20th and 21st centuries. Hum. Reprod. Update 2022, 29, 157–176. [Google Scholar]
  3. Hanson, B.; Eisenberg, M.; Hotaling, J. Male infertility: A biomarker of individual and familial cancer risk. Fertil. Steril. 2018, 109, 6–19. [Google Scholar] [CrossRef] [PubMed]
  4. Del Giudice, F.; Kasman, A.M.; Li, S.; Belladelli, F.; Ferro, M.; de Cobelli, O.; De Berardinis, E.; Busetto, G.M.; Eisenberg, M.L. Increased Mortality Among Men Diagnosed With Impaired Fertility: Analysis of US Claims Data. Urology 2021, 147, 143–149. [Google Scholar] [CrossRef]
  5. Ferlin, A.; Garolla, A.; Ghezzi, M.; Selice, R.; Palego, P.; Caretta, N.; Di Mambro, A.; Valente, U.; De Rocco Ponce, M.; Dipresa, S.; et al. Sperm Count and Hypogonadism as Markers of General Male Health. Eur. Urol. Focus 2021, 7, 205–213. [Google Scholar] [CrossRef]
  6. Glazer, C.H.; Eisenberg, M.L.; Tottenborg, S.S.; Giwercman, A.; Flachs, E.M.; Brauner, E.V.; Vassard, D.; Pinborg, A.; Schmidt, L.; Bonde, J.P. Male factor infertility and risk of death: A nationwide record-linkage study. Hum. Reprod. 2019, 34, 2266–2273. [Google Scholar]
  7. Del Giudice, F.; Kasman, A.M.; Chen, T.; De Berardinis, E.; Busetto, G.M.; Sciarra, A.; Ferro, M.; Lucarelli, G.; Belladelli, F.; Salonia, A.; et al. The Association between Mortality and Male Infertility: Systematic Review and Meta-analysis. Urology 2021, 154, 148–157. [Google Scholar] [CrossRef]
  8. Skakkebaek, N.E.; Rajpert-De Meyts, E.; Louis, G.M.B.; Toppari, J.; Andersson, A.M.; Eisenberg, M.L.; Jensen, T.K.; Jorgensen, N.; Swan, S.H.; Sapra, K.J.; et al. Male Reproductive Disorders and Fertility Trends: Influences of Environment and Genetic Susceptibility. Physiol. Rev. 2016, 96, 55–97. [Google Scholar]
  9. Delli Muti, N.; Finocchi, F.; Tossetta, G.; Salvio, G.; Cutini, M.; Marzioni, D.; Balercia, G. Could SARS-CoV-2 infection affect male fertility and sexuality? Apmis 2022, 130, 243–252. [Google Scholar]
  10. Eisenberg, M.L.; Esteves, S.C.; Lamb, D.J.; Hotaling, J.M.; Giwercman, A.; Hwang, K.; Cheng, Y.S. Male infertility. Nat. Rev. Dis. Primers 2023, 9, 49. [Google Scholar] [CrossRef]
  11. Sharma, A.; Minhas, S.; Dhillo, W.S.; Jayasena, C.N. Male infertility due to testicular disorders. J. Clin. Endocrinol. Metab. 2021, 106, e442–e459. [Google Scholar]
  12. Hoffmann, K.; Gopal, M. Paediatric acute epididymo-orchitis temporally related to SARS-CoV-2 infection: A case series and review of the literature. J. Pediatr. Urol. 2024, 20, 91–94. [Google Scholar] [CrossRef] [PubMed]
  13. Krausz, C. Male infertility: Pathogenesis and clinical diagnosis. Best. Pr. Res. Clin. Endocrinol. Metab. 2011, 25, 271–285. [Google Scholar] [CrossRef] [PubMed]
  14. Shen, H.P.; Song, J.Y.; Zhou, X.; Liu, Y.H.; Chen, Y.J.; Cai, Y.L.; Zhang, Y.B.; Yu, Y.; Chen, X.Q. Exploring the mechanisms of ferroptosis in non-obstructive azoospermia based on bioinformatics and machine learning. Zhonghua Nan Ke Xue 2023, 29, 874–880. [Google Scholar]
  15. Hao, X.; Wang, H.; Cui, F.; Yang, Z.; Ye, L.; Huang, R.; Meng, J. Reduction of SLC7A11 and GPX4 Contributing to Ferroptosis in Sperm from Asthenozoospermia Individuals. Reprod. Sci. 2023, 30, 247–257. [Google Scholar] [CrossRef]
  16. Ammar, O.; Tekeya, O.; Hannachi, I.; Sallem, A.; Haouas, Z.; Mehdi, M. Increased Sperm DNA Fragmentation in Infertile Men with Varicocele: Relationship with Apoptosis, Seminal Oxidative Stress, and Spermatic Parameters. Reprod. Sci. 2021, 28, 909–919. [Google Scholar] [CrossRef]
  17. Sasson, D.C.; Kashanian, J.A. Varicoceles. JAMA 2020, 323, 2210. [Google Scholar] [CrossRef]
  18. Condorelli, R.A.; La Vignera, S.; Mongioi, L.M.; Alamo, A.; Calogero, A.E. Diabetes Mellitus and Infertility: Different Pathophysiological Effects in Type 1 and Type 2 on Sperm Function. Front. Endocrinol. 2018, 9, 268. [Google Scholar] [CrossRef]
  19. Pan, C.; Kong, X.; Wu, Z.; Fei, Q. The role of hepatitis B virus surface protein in inducing Sertoli cell ferroptosis. Andrology 2024, 12, 643–654. [Google Scholar] [CrossRef]
  20. Zhou, L.; Li, L.; Hao, G.; Li, B.; Yang, S.; Wang, N.; Liang, J.; Sun, H.; Ma, S.; Yan, L.; et al. Sperm mtDNA copy number, telomere length, and seminal spermatogenic cells in relation to ambient air pollution: Results of a cross-sectional study in Jing-Jin-Ji region of China. J. Hazard. Mater. 2021, 406, 124308. [Google Scholar] [CrossRef]
  21. Ou, Z.; Wen, Q.; Deng, Y.; Yu, Y.; Chen, Z.; Sun, L. Cigarette smoking is associated with high level of ferroptosis in seminal plasma and affects semen quality. Reprod. Biol. Endocrinol. 2020, 18, 55. [Google Scholar] [CrossRef]
  22. Li, L.; Pei, Z.; Wu, R.; Zhang, Y.; Pang, Y.; Hu, H.; Hu, W.; Geng, Z.; Feng, T.; Niu, Y.; et al. FDX1 regulates leydig cell ferroptosis mediates PM(2.5)-induced testicular dysfunction of mice. Ecotoxicol. Environ. Saf. 2023, 263, 115309. [Google Scholar] [CrossRef] [PubMed]
  23. Zhao, Y.; Zhang, H.; Cui, J.G.; Wang, J.X.; Chen, M.S.; Wang, H.R.; Li, X.N.; Li, J.L. Ferroptosis is critical for phthalates driving the blood-testis barrier dysfunction via targeting transferrin receptor. Redox Biol. 2023, 59, 102584. [Google Scholar] [CrossRef] [PubMed]
  24. Cheng, J.; Yang, L.; Zhang, Z.; Xu, D.; Hua, R.; Chen, H.; Li, X.; Duan, J.; Li, Q. Diquat causes mouse testis injury through inducing heme oxygenase-1-mediated ferroptosis in spermatogonia. Ecotoxicol. Environ. Saf. 2024, 280, 116562. [Google Scholar] [CrossRef]
  25. Liu, X.; Ai, Y.; Xiao, M.; Wang, C.; Shu, Z.; Yin, J.; Chu, Y.; Xiao, Q.; Liu, B. PM 2.5 juvenile exposure-induced spermatogenesis dysfunction by triggering testes ferroptosis and antioxidative vitamins intervention in adult male rats. Environ. Sci. Pollut. Res. Int. 2023, 30, 111051–111061. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, C.; Liu, X.; Shu, Z.; Yin, J.; Xiao, M.; Ai, Y.; Zhao, P.; Luo, Z.; Liu, B. Exposure to automobile exhaust-derived PM2.5 induces spermatogenesis dysfunction by damaging UPR(mt) of prepubertal rats. Ecotoxicol. Environ. Saf. 2022, 245, 114087. [Google Scholar] [CrossRef]
  27. Huang, X.; Fu, Y.; Wang, S.; Guo, Q.; Wu, Y.; Zheng, X.; Wang, J.; Wu, S.; Shen, L.; Wei, G. 2,2′,4,4′-Tetrabromodiphenyl ether exposure disrupts blood-testis barrier integrity through CMA-mediated ferroptosis. Sci. Total Environ. 2024, 948, 174738. [Google Scholar] [CrossRef]
  28. Ma, T.; Cheng, H.; Kong, L.; Shen, C.; Jin, H.; Li, H.; Pan, C.; Liang, J. Combined exposure of PS-MPs with NaF induces Sertoli cell death and dysfunction via ferroptosis and apoptosis. Toxicology 2024, 506, 153849. [Google Scholar] [CrossRef]
  29. Wang, Y.; Wu, J.; Zhang, M.; OuYang, H.; Li, M.; Jia, D.; Wang, R.; Zhou, W.; Liu, H.; Hu, Y.; et al. Cadmium exposure during puberty damages testicular development and spermatogenesis via ferroptosis caused by intracellular iron overload and oxidative stress in mice. Environ. Pollut. 2023, 325, 121434. [Google Scholar] [CrossRef]
  30. Zeng, Q.; Yi, H.; Huang, L.; An, Q.; Wang, H. Long-term arsenite exposure induces testicular toxicity by redox imbalance, G2/M cell arrest and apoptosis in mice. Toxicology 2019, 411, 122–132. [Google Scholar] [CrossRef]
  31. Zhao, Y.; Zhu, Q.; Lin, J.; Cai, J. Association of Exposure to Particulate Matter Air Pollution With Semen Quality Among Men in China. JAMA Netw. Open 2022, 5, e2148684. [Google Scholar] [CrossRef]
  32. Hammoud, A.; Carrell, D.T.; Gibson, M.; Sanderson, M.; Parker-Jones, K.; Peterson, C.M. Decreased sperm motility is associated with air pollution in Salt Lake City. Fertil. Steril. 2010, 93, 1875–1879. [Google Scholar] [CrossRef] [PubMed]
  33. Mao, Z.; Ji, Q.; Chen, P.; Zhong, K.; Zeng, X. Hydrogen sulfide protects against toxicant acrolein-induced ferroptotic cell death in Sertoli cells. Front. Pharmacol. 2024, 15, 1440147. [Google Scholar] [CrossRef]
  34. Schiavoni, V.; Emanuelli, M.; Milanese, G.; Galosi, A.B.; Pompei, V.; Salvolini, E.; Campagna, R. Nrf2 Signaling in Renal Cell Carcinoma: A Potential Candidate for the Development of Novel Therapeutic Strategies. Int. J. Mol. Sci. 2024, 25, 13239. [Google Scholar] [CrossRef] [PubMed]
  35. Mancuso, F.; Arato, I.; Bellucci, C.; Lilli, C.; Eugeni, E.; Aglietti, M.C.; Stabile, A.M.; Pistilli, A.; Brancorsini, S.; Gaggia, F.; et al. Zinc restores functionality in porcine prepubertal Sertoli cells exposed to subtoxic cadmium concentration via regulating the Nrf2 signaling pathway. Front. Endocrinol. 2023, 14, 962519. [Google Scholar] [CrossRef] [PubMed]
  36. Pan, C.; Ji, Z.; Wang, Q.; Zhang, Z.; Wang, Z.; Li, C.; Lu, S.; Ge, P. Cuproptosis: Mechanisms, biological significance, and advances in disease treatment-A systematic review. CNS Neurosci. Ther. 2024, 30, e70039. [Google Scholar] [CrossRef]
  37. Parveen, S.; Rizvi, A.; Akhtar, K.; Khan, A.A.; Naseem, I. Nickel-induced oxidative stress causes cell death in testicles: Implications for male infertility. Naunyn Schmiedebergs Arch. Pharmacol. 2024, 397, 1659–1670. [Google Scholar] [CrossRef]
  38. de Ligny, W.; Smits, R.M.; Mackenzie-Proctor, R.; Jordan, V.; Fleischer, K.; de Bruin, J.P.; Showell, M.G. Antioxidants for male subfertility. Cochrane Database Syst. Rev. 2022, 5, CD007411. [Google Scholar]
  39. Du, Y.; Guo, Z. Recent progress in ferroptosis: Inducers and inhibitors. Cell Death Discov. 2022, 8, 501. [Google Scholar] [CrossRef]
  40. Zhao, X.; Liu, Z.; Gao, J.; Li, H.; Wang, X.; Li, Y.; Sun, F. Inhibition of ferroptosis attenuates busulfan-induced oligospermia in mice. Toxicology 2020, 440, 152489. [Google Scholar] [CrossRef]
  41. Xu, J.; Zhang, L.; Si, Y.; Huang, W.; Liu, R.; Liu, Z.; Jiang, Z.; Xu, F. Ferritinophagy-mediated ferroptosis of spermatogonia is involved in busulfan-induced oligospermia in the mice. Chem. Biol. Interact. 2024, 390, 110870. [Google Scholar] [CrossRef]
  42. Dong, J.; Dong, Y.; Chen, H.; Ye, T.; Chen, G.; Fan, B.; Wang, X.; Shi, J.; Wang, C. Ursonic acid attenuates spermatogenesis in oligozoospermia mice through inhibiting ferroptosis. Bioorg Chem. 2024, 144, 107174. [Google Scholar]
  43. Ma, B.; Zhang, J.; Zhu, Z.; Bao, X.; Zhang, M.; Ren, C.; Zhang, Q. Aucubin, a natural iridoid glucoside, attenuates oxidative stress-induced testis injury by inhibiting JNK and CHOP activation via Nrf2 up-regulation. Phytomedicine 2019, 64, 153057. [Google Scholar] [PubMed]
  44. Kopalli, S.R.; Cha, K.M.; Cho, J.Y.; Kim, S.K.; Koppula, S. Cordycepin mitigates spermatogenic and redox related expression in H(2)O(2)-exposed Leydig cells and regulates testicular oxidative apoptotic signalling in aged rats. Pharm. Biol. 2022, 60, 404–416. [Google Scholar] [CrossRef]
  45. Khalil, A.S.M.; Giribabu, N.; Yelumalai, S.; Shahzad, H.; Kilari, E.K.; Salleh, N. Myristic acid defends against testicular oxidative stress, inflammation, apoptosis: Restoration of spermatogenesis, steroidogenesis in diabetic rats. Life Sci. 2021, 278, 119605. [Google Scholar]
  46. Liu, C.Y.; Chen, C.C.; Chiang, L.H.; Yang, B.H.; Chang, T.C.; Tsao, C.W. Hirsutella sinensis intensifies testicular function and spermatogenesis in male mice with high-fat diet-induced obesity. J. Chin. Med. Assoc. 2024, 87, 765–773. [Google Scholar]
  47. Awny, M.M.; Al-Mokaddem, A.K.; Ali, B.M. Mangiferin mitigates di-(2-ethylhexyl) phthalate-induced testicular injury in rats by modulating oxidative stress-mediated signals, inflammatory cascades, apoptotic pathways, and steroidogenesis. Arch. Biochem. Biophys. 2021, 711, 108982. [Google Scholar]
  48. Zhu, Y.; Zhang, J.; Liu, Q.; Xin, X.; Dong, L.; Wang, B.; Li, H.; Li, D.; Wang, J.; Guan, S.; et al. Semen Cuscutae-Fructus Lycii attenuates tripterygium glycosides-induced spermatogenesis dysfunction by inhibiting oxidative stress-mediated ferroptosis via the Nrf2/HO-1 pathway. Phytomedicine 2024, 135, 156221. [Google Scholar]
  49. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular mechanisms and health implications. Cell Res. 2021, 31, 107–125. [Google Scholar] [CrossRef]
  50. Bravo, A.; Sánchez, R.; Zambrano, F.; Uribe, P. Exogenous Oxidative Stress in Human Spermatozoa Induces Opening of the Mitochondrial Permeability Transition Pore: Effect on Mitochondrial Function, Sperm Motility and Induction of Cell Death. Antioxidants 2024, 13, 739. [Google Scholar] [CrossRef] [PubMed]
  51. Li, J.; Chen, D.; Suo, J.; Li, J.; Zhang, Y.; Wang, Y.; Deng, Z.; Zhang, Q.; Ma, B. Triptolide induced spermatogenesis dysfunction via ferroptosis activation by promoting K63-linked GPX4 polyubiquitination in spermatocytes. Chem. Biol. Interact. 2024, 399, 111130. [Google Scholar]
  52. Mou, Y.; Wang, J.; Wu, J.; He, D.; Zhang, C.; Duan, C.; Li, B. Ferroptosis, a new form of cell death: Opportunities and challenges in cancer. J. Hematol. Oncol. 2019, 12, 34. [Google Scholar] [PubMed]
  53. Liu, Y.; Wan, Y.; Jiang, Y.; Zhang, L.; Cheng, W. GPX4: The hub of lipid oxidation, ferroptosis, disease and treatment. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188890. [Google Scholar]
  54. Zhu, H.; Cheng, Y.; Wang, X.; Yang, X.; Liu, M.; Liu, J.; Liu, S.; Wang, H.; Zhang, A.; Li, R.; et al. Gss deficiency causes age-related fertility impairment via ROS-triggered ferroptosis in the testes of mice. Cell Death Dis. 2023, 14, 845. [Google Scholar] [PubMed]
  55. Nashed, M.S.; Hassanen, E.I.; Issa, M.Y.; Tohamy, A.F.; Prince, A.M.; Hussien, A.M.; Soliman, M.M. The mollifying effect of Sambucus nigra extract on StAR gene expression, oxidative stress, and apoptosis induced by fenpropathrin in male rats. Food Chem. Toxicol. 2024, 189, 114744. [Google Scholar]
  56. Xie, Y.; Kang, R.; Klionsky, D.J.; Tang, D. GPX4 in cell death, autophagy, and disease. Autophagy 2023, 19, 2621–2638. [Google Scholar] [CrossRef]
  57. Jin, Z.; Yang, Y.; Cao, Y.; Wen, Q.; Xi, Y.; Cheng, J.; Zhao, Q.; Weng, J.; Hong, K.; Jiang, H.; et al. The gut metabolite 3-hydroxyphenylacetic acid rejuvenates spermatogenic dysfunction in aged mice through GPX4-mediated ferroptosis. Microbiome 2023, 11, 212. [Google Scholar]
  58. Anandhan, A.; Dodson, M.; Shakya, A.; Chen, J.; Liu, P.; Wei, Y.; Tan, H.; Wang, Q.; Jiang, Z.; Yang, K.; et al. NRF2 controls iron homeostasis and ferroptosis through HERC2 and VAMP8. Sci. Adv. 2023, 9, eade9585. [Google Scholar] [CrossRef]
  59. Han, P.; Wang, X.; Zhou, T.; Cheng, J.; Wang, C.; Sun, F.; Zhao, X. Inhibition of ferroptosis attenuates oligospermia in male Nrf2 knockout mice. Free Radic. Biol. Med. 2022, 193 Pt 1 Pt 1, 421–429. [Google Scholar] [CrossRef]
  60. Yu, B.; Lin, H.; Yang, L.; Chen, K.; Luo, H.; Liu, J.; Gao, X.; Xia, X.; Huang, Z. Genetic variation in the Nrf2 promoter associates with defective spermatogenesis in humans. J. Mol. Med. 2012, 90, 1333–1342. [Google Scholar] [CrossRef]
  61. Aydos, O.S.; Yukselten, Y.; Aydos, D.; Sunguroglu, A.; Aydos, K. Relationship between functional Nrf2 gene promoter polymorphism and sperm DNA damage in male infertility. Syst. Biol. Reprod. Med. 2021, 67, 399–412. [Google Scholar]
  62. Chen, K.; Mai, Z.; Zhou, Y.; Gao, X.; Yu, B. Low NRF2 mRNA expression in spermatozoa from men with low sperm motility. Tohoku J. Exp. Med. 2012, 228, 259–266. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, S.Z.; Liu, J.N.; Zhou, F.F.; Wang, Y.J.; Zhang, P.; Cheng, S.T. Decreased Nrf2 protein level and low sperm quality in intractable spermatocystitis. Asian J. Androl. 2024, 26, 189–194. [Google Scholar] [CrossRef] [PubMed]
  64. Aydemir, B.; Kiziler, A.R.; Onaran, I.; Alici, B.; Ozkara, H.; Akyolcu, M.C. Impact of Cu and Fe concentrations on oxidative damage in male infertility. Biol. Trace Elem. Res. 2006, 112, 193–203. [Google Scholar] [CrossRef]
  65. Chabchoub, I.; Nouioui, M.A.; Araoud, M.; Mabrouk, M.; Amira, D.; Ben Aribia, M.H.; Mahmoud, K.; Zhioua, F.; Merdassi, G.; Hedhili, A. Effects of lead, cadmium, copper and zinc levels on the male reproductive function. Andrologia 2021, 53, e14181. [Google Scholar] [CrossRef] [PubMed]
  66. Yin, T.; Yue, X.; Li, Q.; Zhou, X.; Dong, R.; Chen, J.; Zhang, R.; Wang, X.; He, S.; Jiang, T.; et al. The Association Between the Levels of Oxidative Stress Indicators (MDA, SOD, and GSH) in Seminal Plasma and the Risk of Idiopathic Oligo-asthenotera-tozoospermia: Does Cu or Se Level Alter the Association? Biol. Trace Elem. Res. 2024, 202, 2941–2953. [Google Scholar] [CrossRef]
  67. Akinloye, O.; Abbiyesuku, F.M.; Oguntibeju, O.O.; Arowojolu, A.O.; Truter, E.J. The impact of blood and seminal plasma zinc and copper concentrations on spermogram and hormonal changes in infertile Nigerian men. Reprod. Biol. 2011, 11, 83–98. [Google Scholar] [CrossRef]
  68. Chen, L.; Min, J.; Wang, F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct. Target. Ther. 2022, 7, 378. [Google Scholar] [CrossRef]
  69. Al-Musawi, M.M.S.; Al-Shmgani, H.; Al-Bairuty, G.A. Histopathological and Biochemical Comparative Study of Copper Oxide Nanoparticles and Copper Sulphate Toxicity in Male Albino Mice Reproductive System. Int. J. Biomater. 2022, 2022, 4877637. [Google Scholar] [CrossRef]
  70. Nicy, V.; Das, M.; Gurusubramanian, G.; Mondal, P.; Roy, V.K. Treatment of copper nanoparticles (CuNPs) for two spermatogenic cycles impairs testicular activity via down-regulating steroid receptors and inhibition of germ cell proliferation in a mice model. Nanotoxicology 2022, 16, 658–678. [Google Scholar] [CrossRef]
  71. Liu, J.Y.; Yang, X.; Sun, X.D.; Zhuang, C.C.; Xu, F.B.; Li, Y.F. Suppressive Effects of Copper Sulfate Accumulation on the Spermatogenesis of Rats. Biol. Trace Elem. Res. 2016, 174, 356–361. [Google Scholar] [CrossRef]
  72. Nicy, V.; Gurusubramanian, G.; Roy, V.K. Effects of chronic CuNPs treatment followed by termination for two spermatogenic cycles in the testicular functions of mice. Reprod. Toxicol. 2024, 129, 108669. [Google Scholar] [CrossRef]
  73. Zhang, J.Y.; Yu, X.J.; Li, J.J.; Xiao, Y.; Li, G.S.; Yang, F.; Dong, L. Cuproptosis mediates copper-induced testicular spermatogenic cell death. Asian J. Androl. 2024, 26, 295–301. [Google Scholar] [CrossRef]
  74. Guo, H.; Ouyang, Y.; Wang, J.; Cui, H.; Deng, H.; Zhong, X.; Jian, Z.; Liu, H.; Fang, J.; Zuo, Z.; et al. Cu-induced spermatogenesis disease is related to oxidative stress-mediated germ cell apoptosis and DNA damage. J. Hazard. Mater. 2021, 416, 125903. [Google Scholar] [CrossRef]
  75. Li, Y.; Chen, H.; Liao, J.; Chen, K.; Javed, M.T.; Qiao, N.; Zeng, Q.; Liu, B.; Yi, J.; Tang, Z.; et al. Long-term copper exposure promotes apoptosis and autophagy by inducing oxidative stress in pig testis. Environ. Sci. Pollut. Res. Int. 2021, 28, 55140–55153. [Google Scholar] [CrossRef]
  76. Zhao, M.; Yu, W.X.; Liu, S.J.; Deng, Y.J.; Zhao, Z.W.; Guo, J.; Gao, Q.H. Identification and immuno-infiltration analysis of cuproptosis regulators in human spermatogenic dysfunction. Front. Genet. 2023, 14, 1115669. [Google Scholar] [CrossRef]
  77. Zhao, D.; Wu, L.; Fang, X.; Wang, L.; Liu, Q.; Jiang, P.; Ji, Z.; Zhang, N.; Yin, M.; Han, H. Copper exposure induces inflammation and PANoptosis through the TLR4/NF-κB signaling pathway, leading to testicular damage and impaired spermatogenesis in Wilson disease. Chem. Biol. Interact. 2024, 396, 111060. [Google Scholar] [CrossRef]
  78. Shaoyong, W.; Wang, W.; Pan, B.; Liu, R.; Yin, L.; Wangjie, R.; Tian, H.; Wang, Y.; Jin, M. Transgenerational Inheritance Effects of Copper Oxide Nanoparticles (CuONPs) Induced Asthenospermia and Infertility via Gamete H3K9me3 Insufficiency Pathway in Mice. ACS Nano 2024, 18, 20541–20555. [Google Scholar] [CrossRef]
  79. Huang, Q.; Li, J.; Qi, Y.; He, X.; Shen, C.; Wang, C.; Wang, X.; Xia, Q.; Zhang, Y.; Pan, Z.; et al. Copper overload exacerbates testicular aging mediated by lncRNA:CR43306 deficiency through ferroptosis in Drosophila. Redox Biol. 2024, 76, 103315. [Google Scholar] [CrossRef]
  80. Chen, K.; Wu, L.; Liu, Q.; Tan, F.; Wang, L.; Zhao, D.; Fang, X.; Liu, X.; Liu, J.; Han, H. Glutathione improves testicular spermatogenesis through inhibiting oxidative stress, mitochondrial damage, and apoptosis induced by copper deposition in mice with Wilson disease. Biomed. Pharmacother. 2023, 158, 114107. [Google Scholar] [CrossRef]
  81. Yi, H.; Chen, T.; He, G.; Liu, L.; Zhao, J.; Guo, K.; Cao, Y.; Sun, P.; Zhou, X.; Zhang, B.; et al. Retinoic acid mitigates the NSC319726-induced spermatogenesis dysfunction through cuproptosis-independent mechanisms. Cell Biol. Toxicol. 2024, 40, 26. [Google Scholar] [CrossRef]
  82. Seven, I.; Tatli Seven, P.; Gul Baykalir, B.; Parlak Ak, T.; Ozer Kaya, S.; Yaman, M. Bee glue (propolis) improves reproductive organs, sperm quality and histological changes and antioxidant parameters of testis tissues in rats exposed to excess copper. Andrologia 2020, 52, e13540. [Google Scholar] [CrossRef]
  83. Arafa, M.H.; Amin, D.M.; Samir, G.M.; Atteia, H.H. Protective effects of tribulus terrestris extract and angiotensin blockers on testis steroidogenesis in copper overloaded rats. Ecotoxicol. Environ. Saf. 2019, 178, 113–122. [Google Scholar] [CrossRef]
  84. Wang, T.; Wu, L.; Chen, Q.; Chen, K.; Tan, F.; Liu, J.; Liu, X.; Han, H. Copper deposition in Wilson’s disease causes male fertility decline by impairing reproductive hormone release through inducing apoptosis and inhibiting ERK signal in hypothalamic-pituitary of mice. Front. Endocrinol. 2022, 13, 961748. [Google Scholar] [CrossRef]
  85. Iorio, G.G.; Conforti, A.; Vallone, R.; Carbone, L.; Matarazzo, M.; De Rosa, A.; De Rosa, P.; Picarelli, S.; Fedele, F.; Perruolo, G.; et al. Reproductive function of long-term treated patients with hepatic onset of Wilson’s disease: A prospective study. Reprod. Biomed. Online 2021, 42, 835–841. [Google Scholar] [CrossRef]
  86. Wang, X.; Zhou, M.; Liu, Y.; Si, Z. Cope with copper: From copper linked mechanisms to copper-based clinical cancer therapies. Cancer Lett. 2023, 561, 216157. [Google Scholar] [CrossRef]
  87. Kirk, F.T.; Munk, D.E.; Swenson, E.S.; Quicquaro, A.M.; Vendelbo, M.H.; Larsen, A.; Schilsky, M.L.; Ott, P.; Sandahl, T.D. Effects of tetrathiomolybdate on copper metabolism in healthy volunteers and in patients with Wilson disease. J. Hepatol. 2024, 80, 586–595. [Google Scholar] [CrossRef]
  88. Chan, N.; Willis, A.; Kornhauser, N.; Ward, M.M.; Lee, S.B.; Nackos, E.; Seo, B.R.; Chuang, E.; Cigler, T.; Moore, A.; et al. Influencing the Tumor Microenvironment: A Phase II Study of Copper Depletion Using Tetrathiomolybdate in Patients with Breast Cancer at High Risk for Recurrence and in Preclinical Models of Lung Metastases. Clin. Cancer Res. 2017, 23, 666–676. [Google Scholar] [CrossRef] [PubMed]
  89. Ogórek, M.; Herman, S.; Pierzchała, O.; Bednarz, A.; Rajfur, Z.; Baster, Z.; Grzmil, P.; Starzyński, R.R.; Szudzik, M.; Jończy, A.; et al. Molecular machinery providing copper bioavailability for spermatozoa along the epididymial tubule in mouse. Biol. Reprod. 2019, 100, 1505–1520. [Google Scholar] [CrossRef]
  90. Ghaffari, R.; Di Bona, K.R.; Riley, C.L.; Richburg, J.H. Copper transporter 1 (CTR1) expression by mouse testicular germ cells, but not Sertoli cells, is essential for functional spermatogenesis. PLoS ONE 2019, 14, e0215522. [Google Scholar] [CrossRef]
  91. Ogórek, M.; Lenartowicz, M.; Starzyński, R.; Jończy, A.; Staroń, R.; Doniec, A.; Krzeptowski, W.; Bednarz, A.; Pierzchała, O.; Lipiński, P.; et al. Atp7a and Atp7b regulate copper homeostasis in developing male germ cells in mice. Metallomics 2017, 9, 1288–1303. [Google Scholar] [CrossRef]
  92. Zhang, G.; Shen, L.; Li, Z.; Zhao, Y. FDX1 serves as a prognostic biomarker and promotes glioma progression by regulating the immune response. Aging 2023, 15, 4963–4985. [Google Scholar]
  93. Yang, L.; Zhang, Y.; Wang, Y.; Jiang, P.; Liu, F.; Feng, N. Ferredoxin 1 is a cuproptosis-key gene responsible for tumor immunity and drug sensitivity: A pan-cancer analysis. Front. Pharmacol. 2022, 13, 938134. [Google Scholar] [CrossRef]
  94. Jiang, S.; Wei, Y.; Li, Y.; Liu, W.; Wang, Z.; Meng, X.; Zhu, Q.; Shen, L. A comparative cross-platform analysis of cuproptosis-related genes in human nonobstructive azoospermia: An observational study. Medicine 2024, 103, e39176. [Google Scholar]
  95. Gao, W.; Wang, X.; Zhou, Y.; Wang, X.; Yu, Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct. Target. Ther. 2022, 7, 196. [Google Scholar] [CrossRef]
  96. Yamamoto, H.; Zhang, S.; Mizushima, N. Autophagy genes in biology and disease. Nat. Rev. Genet. 2023, 24, 382–400. [Google Scholar]
  97. Qiao, L.; Hu, J.; Qiu, X.; Wang, C.; Peng, J.; Zhang, C.; Zhang, M.; Lu, H.; Chen, W. LAMP2A, LAMP2B and LAMP2C: Similar structures, divergent roles. Autophagy 2023, 19, 2837–2852. [Google Scholar] [CrossRef]
  98. Zhang, G.; Jiang, C.; Yang, Y.; Wang, Y.; Zhou, H.; Dai, S.; Liu, M.; Yang, Y.; Yang, L.; Shen, Q.; et al. Deficiency of cancer/testis antigen gene CT55 causes male infertility in humans and mice. Cell Death Differ. 2023, 30, 500–514. [Google Scholar]
  99. Wang, Y.; Tian, C.C.; Jiao, Y.Y.; Liu, M.R.; Ma, X.S.; Jin, H.X.; Su, Y.C.; Zhang, X.Y.; Niu, W.B.; Yao, G.D.; et al. miR-188-3p-targeted regulation of ATG7 affects cell autophagy in patients with nonobstructive azoospermia. Reprod. Biol. Endocrinol. 2022, 20, 90. [Google Scholar]
  100. Foroozan-Broojeni, S.; Tavalaee, M.; Lockshin, R.A.; Zakeri, Z.; Abbasi, H.; Nasr-Esfahani, M.H. Comparison of main molecular markers involved in autophagy and apoptosis pathways between spermatozoa of infertile men with varicocele and fertile individuals. Andrologia 2019, 51, e13177. [Google Scholar]
  101. Wu, C.; Zhu, S.M.; Cheng, F.; Zhou, X.J.; Yu, W.M.; Rao, T.; Li, H.Y.; Ning, J.Z.; Xia, Y.Q.; Zhu, C.C. Effect of testicular autophagy on spermatogenic cells in varicocele rats. Zhonghua Nan Ke Xue 2020, 26, 111–117. [Google Scholar]
  102. Liu, X.; Xi, H.; Han, S.; Zhang, H.; Hu, J. Zearalenone induces oxidative stress and autophagy in goat Sertoli cells. Ecotoxicol. Environ. Saf. 2023, 252, 114571. [Google Scholar]
  103. Huang, W.; Cao, Z.; Zhang, J.; Ji, Q.; Li, Y. Aflatoxin B1 promotes autophagy associated with oxidative stress-related PI3K/AKT/mTOR signaling pathway in mice testis. Environ. Pollut. 2019, 255, 113317. [Google Scholar] [CrossRef]
  104. Ali, W.; Deng, K.; Sun, J.; Ma, Y.; Liu, Z.; Zou, H. A new insight of cadmium-induced cellular evidence of autophagic-associated spermiophagy during spermatogenesis. Environ. Sci. Pollut. Res. Int. 2023, 30, 101064–101074. [Google Scholar]
  105. Chen, N.; Wan, X.; Wang, M.; Li, Y.; Wang, X.; Zeng, L.; Zhou, J.; Zhang, Y.; Cheng, S.; Shen, Y. Cross-talk between Vimentin and autophagy regulates blood-testis barrier disruption induced by cadmium. Environ. Pollut. 2024, 346, 123625. [Google Scholar]
  106. Guo, H.; Ouyang, Y.; Yin, H.; Cui, H.; Deng, H.; Liu, H.; Jian, Z.; Fang, J.; Zuo, Z.; Wang, X.; et al. Induction of autophagy via the ROS-dependent AMPK-mTOR pathway protects copper-induced spermatogenesis disorder. Redox Biol. 2022, 49, 102227. [Google Scholar] [CrossRef]
  107. Yang, D.; Zhang, M.; Gan, Y.; Yang, S.; Wang, J.; Yu, M.; Wei, J.; Chen, J. Involvement of oxidative stress in ZnO NPs-induced apoptosis and autophagy of mouse GC-1 spg cells. Ecotoxicol. Environ. Saf. 2020, 202, 110960. [Google Scholar]
  108. Hong, Y.; Zhou, X.; Li, Q.; Chen, J.; Wei, Y.; Long, C.; Shen, L.; Zheng, X.; Li, D.; Wang, X.; et al. X-box binding protein 1 caused an imbalance in pyroptosis and mitophagy in immature rats with di-(2-ethylhexyl) phthalate-induced testis toxicity. Genes Dis. 2024, 11, 935–951. [Google Scholar]
  109. Chen, P.; Chen, J.; Zhang, W.; Tang, L.; Cheng, G.; Li, H.; Fan, T.; Wang, J.; Zhong, W.; Song, Y. Biochemical mechanisms of tributyltin chloride-induced cell toxicity in Sertoli cells. Ecotoxicol. Environ. Saf. 2023, 255, 114725. [Google Scholar]
  110. Yang, Y.; Fu, G.; Zhao, X.; Wu, X.; Zhu, K.; Liu, S.; Yuan, D.; Wu, J.; Wang, T.; Zhang, C.; et al. Perfluorooctanoic acid induces tight junction injury of Sertoli cells by blocking autophagic flux. Food Chem. Toxicol. 2023, 173, 113649. [Google Scholar]
  111. Horibe, A.; Eid, N.; Ito, Y.; Otsuki, Y.; Kondo, Y. Ethanol-Induced Autophagy in Sertoli Cells Is Specifically Marked at Androgen-Dependent Stages of the Spermatogenic Cycle: Potential Mechanisms and Implications. Int. J. Mol. Sci. 2019, 20, 184. [Google Scholar] [CrossRef]
  112. Li, G.; Xu, Y.; Li, Y.; Chang, D.; Zhang, P.; Ma, Z.; Chen, D.A.; You, Y.; Huang, X.; Cai, J. Qiangjing tablets ameliorate asthenozoospermia via mitochondrial ubiquitination and mitophagy mediated by LKB1/AMPK/ULK1 signaling. Pharm. Biol. 2023, 61, 271–280. [Google Scholar] [PubMed]
  113. Abu-Baih, R.H.; Abu-Baih, D.H.; Abdel-Hafez, S.M.N.; Fathy, M. Activation of SIRT1/Nrf2/HO-1 and Beclin-1/AMPK/mTOR autophagy pathways by eprosartan ameliorates testicular dysfunction induced by testicular torsion in rats. Sci. Rep. 2024, 14, 12566. [Google Scholar] [CrossRef] [PubMed]
  114. Mostafa, T.; El-Taweel, A.E.I.; Rashed, L.A.; Mohammed, N.A.M.; Akl, E.M. Assessment of seminal cystatin C levels in infertile men with varicocele: A preliminary study. Andrologia 2022, 54, e14278. [Google Scholar]
  115. Xia, Y.; Tian, Y.; Zhou, D.; Zhang, L.; Cai, Y.; Fu, S.; Zhang, X.; Gao, Y.; Chen, Q.; Gao, P. Gut microbiota involved in spermatogenic function of Sancai Lianmei granules in obese mice. Naunyn Schmiedebergs Arch. Pharmacol. 2023, 396, 83–97. [Google Scholar] [CrossRef]
  116. Cao, C.; Zhang, H.; He, Z.; Zhang, K.; Qian, Z.; Shen, J.; Zheng, L.; Xue, M.; Sun, S.; Li, C.; et al. Octanoic acid mitigates busulfan-induced blood-testis barrier damage by alleviating oxidative stress and autophagy. Lipids Health Dis. 2024, 23, 180. [Google Scholar]
  117. Kankılıç, N.A.; Şimşek, H.; Akaras, N.; Gür, C.; İleritürk, M.; Küçükler, S.; Akarsu, S.A.; Kandemir, F.M. Protective effects of naringin on colistin-induced damage in rat testicular tissue: Modulating the levels of Nrf-2/HO-1, AKT-2/FOXO1A, Bax/Bcl2/Caspase-3, and Beclin-1/LC3A/LC3B signaling pathways. J. Biochem. Mol. Toxicol. 2024, 38, e23643. [Google Scholar]
  118. Shojaedini, M.; Hemadi, M.; Saki, G.; Fakhredini, F.; Khodayar, M.J.; Khorsandi, L. Thymoquinone effects on autophagy, apoptosis, and oxidative stress in cisplatin-induced testicular damage in mice. J. Assist. Reprod. Genet. 2024, 41, 1881–1891. [Google Scholar] [CrossRef]
  119. He, H.; Chen, X.; Li, X.; Yang, K.; Li, J.; Shi, H. Lactoferrin alleviates spermatogenesis dysfunction caused by bisphenol A and cadmium via ameliorating disordered autophagy, apoptosis and oxidative stress. Int. J. Biol. Macromol. 2022, 222 Pt A, 1048–1062. [Google Scholar] [CrossRef]
  120. Chen, Z.; Mo, J.; Yang, Q.; Guo, Z.; Li, X.; Xie, D.; Deng, C. MSC-derived exosomes mitigate cadmium-induced male reproductive injury by ameliorating DNA damage and autophagic flux. Ecotoxicol. Environ. Saf. 2024, 276, 116306. [Google Scholar]
  121. Helal, A.M.; Yossef, M.M.; Seif, I.K.; Abd El-Salam, M.; El Demellawy, M.A.; Abdulmalek, S.A.; Ghareeb, A.Z.; Holail, J.; Mohsen Al-Mahallawi, A.; El-Zahaby, S.A.; et al. Nanostructured biloalbuminosomes loaded with berberine and berberrubine for Alleviating heavy Metal-Induced male infertility in rats. Int. J. Pharm. 2024, 667 Pt A, 124892. [Google Scholar]
  122. Wang, H.; Yang, F.; Ye, J.; Dai, X.; Liao, H.; Xing, C.; Jiang, Z.; Peng, C.; Gao, F.; Cao, H. Ginkgo biloba extract alleviates deltamethrin-induced testicular injury by upregulating SKP2 and inhibiting Beclin1-independent autophagy. Phytomedicine 2024, 135, 156245. [Google Scholar] [CrossRef] [PubMed]
  123. Miao, H.; Miao, C.X.; Li, N.; Han, J. FOXJ2 controls meiosis during spermatogenesis in male mice. Mol. Reprod. Dev. 2016, 83, 684–691. [Google Scholar] [CrossRef] [PubMed]
  124. Bai, F.R.; Wu, Q.Q.; Wu, Y.J.; Hu, Y.Q.; Jiang, Z.X.; Lv, H.; Qian, W.Z.; Cai, C.; Wu, J.W. Germline FOXJ2 overexpression causes male infertility via aberrant autophagy activation by LAMP2A upregulation. Cell Death Dis. 2022, 13, 665. [Google Scholar] [CrossRef]
  125. Brower, J.V.; Rodic, N.; Seki, T.; Jorgensen, M.; Fliess, N.; Yachnis, A.T.; McCarrey, J.R.; Oh, S.P.; Terada, N. Evolutionarily conserved mammalian adenine nucleotide translocase 4 is essential for spermatogenesis. J. Biol. Chem. 2007, 282, 29658–29666. [Google Scholar] [CrossRef]
  126. Yang, F.; Yang, X.; Zhu, H.; Wang, X.; Liao, X.; Fu, Y.; Fu, T.; Chen, X.; Sysa, A.; Lyu, J.; et al. The essential role of adenine nucleotide translocase 4 on male reproductive function in mice. Braz. J. Med. Biol. Res. 2024, 57, e13590. [Google Scholar] [CrossRef]
  127. Huang, Q.; Liu, Y.; Zhang, S.; Yap, Y.T.; Li, W.; Zhang, D.; Gardner, A.; Zhang, L.; Song, S.; Hess, R.A.; et al. Autophagy core protein ATG5 is required for elongating spermatid development, sperm individualization and normal fertility in male mice. Autophagy 2021, 17, 1753–1767. [Google Scholar] [CrossRef]
  128. Ke, L.; Lin, X.; Luo, Y.; Tao, S.; Yan, C.; He, Y.; Wu, Y.; Liu, N.; Qin, Y. Autophagy core protein BECN1 is vital for spermatogenesis and male fertility in mice†. Biol. Reprod. 2024, 110, 599–614. [Google Scholar] [CrossRef]
  129. Wang, F.; Trosdal, E.S.; Paddar, M.A.; Duque, T.L.A.; Allers, L.; Mudd, M.; Akepati, P.R.; Javed, R.; Jia, J.; Salemi, M.; et al. The role of ATG5 beyond Atg8ylation and autophagy. Autophagy 2024, 20, 448–450. [Google Scholar] [CrossRef]
  130. Wen, Z.; Zhu, H.; Wu, B.; Zhang, A.; Wang, H.; Cheng, Y.; Zhao, H.; Li, J.; Liu, M.; Gao, J. Cathepsin B plays a role in spermatogenesis and sperm maturation through regulating autophagy and apoptosis in mice. PeerJ 2022, 10, e14472. [Google Scholar] [CrossRef]
  131. Araujo, T.F.; Cordeiro, A.V.; Vasconcelos, D.A.A.; Vitzel, K.F.; Silva, V.R.R. The role of cathepsin B in autophagy during obesity: A systematic review. Life Sci. 2018, 209, 274–281. [Google Scholar] [CrossRef]
  132. Deng, C.Y.; Lv, M.; Luo, B.H.; Zhao, S.Z.; Mo, Z.C.; Xie, Y.J. The Role of the PI3K/AKT/mTOR Signalling Pathway in Male Reproduction. Curr. Mol. Med. 2021, 21, 539–548. [Google Scholar] [PubMed]
  133. Zhou, Y.; Yan, J.; Qiao, L.; Zeng, J.; Cao, F.; Sheng, X.; Qi, X.; Long, C.; Liu, B.; Wang, X.; et al. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Ameliorate Aging-Induced BTB Impairment in Porcine Testes by Activating Autophagy and Inhibiting ROS/NLRP3 Inflammasomes via the AMPK/mTOR Signaling Pathway. Antioxidants 2024, 13, 183. [Google Scholar] [CrossRef] [PubMed]
  134. Menon, M.B.; Dhamija, S. Beclin 1 Phosphorylation—at the Center of Autophagy Regulation. Front. Cell Dev. Biol. 2018, 6, 137. [Google Scholar] [CrossRef]
  135. Dong, F.; Ma, Y.; Chen, X.F. Identification of a novel pyroptosis-related gene signature in human spermatogenic dysfunction. J. Assist. Reprod. Genet. 2023, 40, 2251–2266. [Google Scholar] [CrossRef]
  136. Jia, Z.C.; Liu, S.J.; Chen, T.F.; Shi, Z.Z.; Li, X.L.; Gao, Z.W.; Zhang, Q.; Zhong, C.F. Chlorogenic acid can improve spermatogenic dysfunction in rats with varicocele by regulating mitochondrial homeostasis and inhibiting the activation of NLRP3 inflammasomes by oxidative mitochondrial DNA and cGAS/STING pathway. Bioorg. Chem. 2024, 150, 107571. [Google Scholar] [CrossRef]
  137. Camargo, M.; Ibrahim, E.; Intasqui, P.; Belardin, L.B.; Antoniassi, M.P.; Lynne, C.M.; Brackett, N.L.; Bertolla, R.P. Seminal inflammasome activity in the adult varicocele. Hum. Fertil. 2022, 25, 548–556. [Google Scholar] [CrossRef]
  138. Elmorsy, E.H.; Aly, R.G.; Badae, N.M.; Aboghazala, M.M.; Omar, S.S. Zinc alleviates high fat diet-induced spermatogenic dysfunction in Wistar rats: Role of oxidative stress, HMGB1 and inflammasome. Rev. Int. Androl. 2024, 22, 44–52. [Google Scholar]
  139. Zhong, Y.; Deng, H.; Zhao, J.; Luo, G.; Li, H. Effects of acetate-producing Blautia wexlerae on oxidative stress and NLRP3 inflammasome in obesity-associated male infertility. Med. Microbiol. Immunol. 2024, 213, 11. [Google Scholar] [CrossRef]
  140. Cheng, A.; Luo, H.; Fan, B.; Xiang, Q.; Nie, Z.; Feng, S.; Qiao, Y.; Wu, Y.; Zhu, Q.; Liu, R.; et al. Fluoride induces pyroptosis via IL-17A-mediated caspase-1/11-dependent pathways and Bifidobacterium intervention in testis. Sci. Total Environ. 2024, 926, 172036. [Google Scholar] [CrossRef]
  141. Zhang, B.; Zhong, Y.; Du, J.; Ye, R.; Fan, B.; Deng, Y.; Bai, R.; Feng, Y.; Yang, X.; Huang, Y.; et al. 1,2-Dichloroethane induces testicular pyroptosis by activating piR-mmu-1019957/IRF7 pathway and the protective effects of melatonin. Environ. Int. 2024, 184, 108480. [Google Scholar] [CrossRef]
  142. Liu, H.; Du, X.; Zhang, Z.; Ge, K.; Chen, X.; Losiewicz, M.D.; Guo, H.; Zhang, H. Co-exposure of microcystin and nitrite enhanced spermatogenic disorders: The role of mtROS-mediated pyroptosis and apoptosis. Environ. Int. 2024, 188, 108771. [Google Scholar] [CrossRef]
  143. Skóra, B.; Piechowiak, T.; Szychowski, K.A. Engagement of specific intracellular pathways in the inflammation-based reprotoxicity effect of small-size silver nanoparticles on spermatogonia and spermatocytes invitro cell models. Chemosphere 2024, 363, 142897. [Google Scholar] [CrossRef]
  144. Zheng, X.; Guo, C.; Lv, Z.; Jiang, H.; Li, S.; Yu, L.; Zhang, Z. From animal to cell model: Pyroptosis targeted-fibrosis is a novel mechanism of lead-induced testicular toxicity. Food Chem. Toxicol. 2023, 178, 113886. [Google Scholar] [CrossRef]
  145. Ouyang, K.W.; Wang, T.T.; Wang, H.; Luo, Y.X.; Hu, Y.F.; Zheng, X.M.; Ling, Q.; Wang, K.W.; Xiong, Y.W.; Zhang, J.; et al. m6A-methylated Lonp1 drives mitochondrial proteostasis stress to induce testicular pyroptosis upon environmental cadmium exposure. Sci. Total Environ. 2024, 931, 172938. [Google Scholar] [CrossRef]
  146. Taha, M.; Ali, L.S.; El-Nablaway, M.; Ibrahim, M.M.; Badawy, A.M.; Farage, A.E.; Ibrahim, H.S.A.; Zaghloul, R.A.; Hussin, E. Multifaceted Impacts of Monosodium Glutamate on Testicular Health: Insights into Pyroptosis and Therapeutic Potential of Resveratrol. Folia Morphol. 2024, 84, 151–166. [Google Scholar] [CrossRef]
  147. Rao, Z.; Zhu, Y.; Yang, P.; Chen, Z.; Xia, Y.; Qiao, C.; Liu, W.; Deng, H.; Li, J.; Ning, P.; et al. Pyroptosis in inflammatory diseases and cancer. Theranostics 2022, 12, 4310–4329. [Google Scholar] [CrossRef]
  148. Coll, R.C.; Schroder, K.; Pelegrín, P. NLRP3 and pyroptosis blockers for treating inflammatory diseases. Trends Pharmacol. Sci. 2022, 43, 653–668. [Google Scholar] [CrossRef]
  149. Miao, R.; Jiang, C.; Chang, W.Y.; Zhang, H.; An, J.; Ho, F.; Chen, P.; Zhang, H.; Junqueira, C.; Amgalan, D.; et al. Gasdermin D permeabilization of mitochondrial inner and outer membranes accelerates and enhances pyroptosis. Immunity 2023, 56, 2523–2541.e8. [Google Scholar] [CrossRef]
  150. Liu, W.; Li, X.; Ma, Q.; Zhu, Y.; Zhao, W.; Yang, Y.; Xiao, W.; Huang, D.; Cai, F.; Chan, D.Y.L.; et al. Testis cell pyroptosis mediated by CASP1 and CASP4: Possible sertoli cell-only syndrome pathogenesis. Reprod. Biol. Endocrinol. 2023, 21, 53. [Google Scholar] [CrossRef]
  151. Ning, J.; Wang, J.; Xu, L.; Li, H. MiR-153 is Involved in Testicular Ischemia/Reperfusion Injury by Directly Targeting FOXO3 and Regulating Spermatogonia Pyroptosis. Mol. Biotechnol. 2024, 67, 1707–1719. [Google Scholar] [CrossRef]
  152. He, K.X.; Xu, L.; Ning, J.Z.; Cheng, F. MiR-195-5p is involved in testicular ischemia/reperfusion injury by directly targeting PELP1 and regulating spermatogonia pyroptosis. Int. Immunopharmacol. 2023, 121, 110427. [Google Scholar] [CrossRef]
  153. Ma, C.; Huang, J.; Jiang, Y.; Liu, L.; Wang, N.; Huang, S.; Li, H.; Zhang, X.; Wen, S.; Wang, B.; et al. Gasdermin D in macrophages drives orchitis by regulating inflammation and antigen presentation processes. EMBO Mol. Med. 2024, 16, 361–385. [Google Scholar] [CrossRef]
  154. Sheng, W.; Xu, W.; Ding, J.; Lu, B.; Liu, L.; He, Q.; Zhou, Q. Guijiajiao (Colla Carapacis et Plastri, CCP) prevents male infertility via gut microbiota modulation. Chin. J. Nat. Med. 2023, 21, 403–410. [Google Scholar] [CrossRef]
  155. Bazrafkan, M.; Hosseini, E.; Nazari, M.; Amorim, C.A.; Sadeghi, M.R. NLRP3 inflammasome: A joint, potential therapeutic target in management of COVID-19 and fertility problems. J. Reprod. Immunol. 2021, 148, 103427. [Google Scholar] [CrossRef]
  156. Al-Madhagi, H.; Muhammed, M.T. Targeting COVID-19 and varicocele by blocking inflammasome: Ligand-based virtual screening. Arch. Biochem. Biophys. 2024, 759, 110107. [Google Scholar]
  157. Hasani, A.; Khosravi, A.; Behnam, P.; Ramezani, F.; Eslami Farsani, B.; Aliaghaei, A.; Pirani, M.; Akaberi-Nasrabadi, S.; Abdi, S.; Abdollahifar, M.A. Non-apoptotic cell death such as pyroptosis, autophagy, necroptosis and ferroptosis acts as partners to induce testicular cell death after scrotal hyperthermia in mice. Andrologia 2022, 54, e14320. [Google Scholar] [CrossRef]
  158. Li, M.Y.; Zhu, X.L.; Zhao, B.X.; Shi, L.; Wang, W.; Hu, W.; Qin, S.L.; Chen, B.H.; Zhou, P.H.; Qiu, B.; et al. Adrenomedullin alleviates the pyroptosis of Leydig cells by promoting autophagy via the ROS-AMPK-mTOR axis. Cell Death Dis. 2019, 10, 489. [Google Scholar]
  159. Zheng, P.; Zhou, C.; Ding, Y.; Duan, S. Disulfidptosis: A new target for metabolic cancer therapy. J. Exp. Clin. Cancer Res. 2023, 42, 103. [Google Scholar] [CrossRef]
  160. Liu, X.; Nie, L.; Zhang, Y.; Yan, Y.; Wang, C.; Colic, M.; Olszewski, K.; Horbath, A.; Chen, X.; Lei, G.; et al. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat. Cell Biol. 2023, 25, 404–414. [Google Scholar] [CrossRef]
  161. Zhuge, R.; Zhang, L.; Xue, Q.; Wang, R.; Xu, J.; Wang, C.; Meng, C.; Lu, R.; Yin, F.; Guo, L. Ferritinophagy is involved in hexavalent chromium-induced ferroptosis in Sertoli cells. Toxicol. Appl. Pharmacol. 2024, 492, 117139. [Google Scholar] [CrossRef]
  162. Yang, H.; Ding, L.; Xu, B.; Zhang, Z.; Dai, W.; He, T.; Liu, L.; Du, X.; Fu, X. Lycium barbarum polysaccharide alleviates ferroptosis in Sertoli cells through NRF2/SLC7A11/GPX4 pathway and ameliorates DEHP-induced male reproductive damage in mice. Int. J. Biol. Macromol. 2024, 282 Pt 5, 137241. [Google Scholar] [CrossRef] [PubMed]
  163. Cao, Y.; Jin, Z.; Xi, Y.; Cheng, J.; Fang, Z.; Zhao, Q.; Weng, J.; Zhu, J.; Tang, Y.; Zhang, Z.; et al. Roles of ferroptosis in type 1 diabetes induced spermatogenic dysfunction. Free Radic. Biol. Med. 2024, 214, 193–205. [Google Scholar] [CrossRef] [PubMed]
  164. Elseweidy, M.M.; Harb, N.G.; Ali, A.A.; El-Aziz, R.M.A.; Elrashidy, R.A. Sulforaphane substantially impedes testicular ferroptosis in adult rats exposed to di-2-ethylhexyl phthalate through activation of NRF-2/SLC7A11/GPX-4 trajectory. Naunyn Schmiedebergs Arch. Pharmacol. 2024, 398, 3163–3175. [Google Scholar] [CrossRef]
  165. Shi, Y.; Yin, L.; Li, J.Y.; Zhou, S.M.; Wang, N.; Chen, H.Q.; Zeng, Y.; Li, Y.W.; Liu, W.B. FTO mediates bisphenol F-induced blood-testis barrier impairment through regulating ferroptosis via YTHDF1/TfRc and YTHDF2/SLC7A11 signal axis. Environ. Pollut. 2024, 359, 124531. [Google Scholar] [CrossRef]
  166. Fan, Z.; Xin, P.; Zhao, L.; Kong, C.; Piao, C.; Wu, Z.; Qiu, Z.; Zhao, W.; Zhang, Z. Ferroptosis Is Crucial for Cisplatin Induced Sertoli Cell Injury via N6-Methyladenosine Dependent Manner. World J. Mens Health 2024, 42, 865–880. [Google Scholar] [CrossRef]
Figure 1. Modes of cell death in the human testis. The schematic illustrates distinct forms of cell death within testicular compartments. Color coding: blue (seminiferous tubules, housing germ cells), red (Sertoli cells), and yellow (Leydig cells). Specific cell death modalities are annotated as follows: a. ferroptosis of human Sertoli cell, b. ferroptosis of human spermatogonial cell, c. ferroptosis of human sperm cell, d. apoptosis of human spermatocyte, e. apoptosis of human Sertoli cell, f. apoptosis of human spermatogonial stem cell, g. apoptosis of human sperm cell, h. apoptosis of human spermatogonial cell, i. apoptosis of human sperm cell, j. pyroptosis of human Leydig cell, k. pyrosis of human spermatogonial cell, l. pyroptosis of human Sertoli cell, m. autophagy of human sperm cell, and n. autophagy of human Sertoli cell. The diagram was drawn using Adobe Illustrator 2023.
Figure 1. Modes of cell death in the human testis. The schematic illustrates distinct forms of cell death within testicular compartments. Color coding: blue (seminiferous tubules, housing germ cells), red (Sertoli cells), and yellow (Leydig cells). Specific cell death modalities are annotated as follows: a. ferroptosis of human Sertoli cell, b. ferroptosis of human spermatogonial cell, c. ferroptosis of human sperm cell, d. apoptosis of human spermatocyte, e. apoptosis of human Sertoli cell, f. apoptosis of human spermatogonial stem cell, g. apoptosis of human sperm cell, h. apoptosis of human spermatogonial cell, i. apoptosis of human sperm cell, j. pyroptosis of human Leydig cell, k. pyrosis of human spermatogonial cell, l. pyroptosis of human Sertoli cell, m. autophagy of human sperm cell, and n. autophagy of human Sertoli cell. The diagram was drawn using Adobe Illustrator 2023.
Ijms 26 03496 g001
Figure 2. Environmental factors cause cell death, inducing male infertility. The diagram illustrates three types of cell death caused by different environmental factors: ferroptosis, pyroptosis, and autophagy. The black arrows indicate that the substances within each box cause a type of cell death. PM2.5 (particulate matter 2.5); DEHP (plasticizer di-(2-ethylhexyl) phthalate); FNP (insecticide 3-fluoro-4-nitrophenol); DQ (herbicide 1,1′-ethyl-2,2′ -bipyridine dibromide); ACR (acrolein); PBDE-47(2,2′,4,4′-tetrabromodiphenyl ether); Cd (cadmium); NaF and MPs (the combined effects of sodium fluoride and microplastics); AgNP (silver nanoparticles); ZnO NPs (zinc oxide nanoparticles); AFB1 (aflatoxin B1); ZEA (zealenone); TBTCL (tributyl tin chloride); PFOA (perfluorooctanoic acid). The diagram was drawn using Adobe Illustrator 2023.
Figure 2. Environmental factors cause cell death, inducing male infertility. The diagram illustrates three types of cell death caused by different environmental factors: ferroptosis, pyroptosis, and autophagy. The black arrows indicate that the substances within each box cause a type of cell death. PM2.5 (particulate matter 2.5); DEHP (plasticizer di-(2-ethylhexyl) phthalate); FNP (insecticide 3-fluoro-4-nitrophenol); DQ (herbicide 1,1′-ethyl-2,2′ -bipyridine dibromide); ACR (acrolein); PBDE-47(2,2′,4,4′-tetrabromodiphenyl ether); Cd (cadmium); NaF and MPs (the combined effects of sodium fluoride and microplastics); AgNP (silver nanoparticles); ZnO NPs (zinc oxide nanoparticles); AFB1 (aflatoxin B1); ZEA (zealenone); TBTCL (tributyl tin chloride); PFOA (perfluorooctanoic acid). The diagram was drawn using Adobe Illustrator 2023.
Ijms 26 03496 g002
Figure 3. Four cell death mechanisms in male infertility. (a). Pyroptosis. Some signaling molecules or stimuli activate pattern recognition receptors, thereby activating the inflammasome, activating caspase-1, cleaving the GSDMD protein, perforating the cell membrane, causing cell death, and releasing inflammatory factors to amplify the inflammatory response. Black arrows indicate activation or upregulation; adaptor protein (ASC); pattern recognition receptors (NLRP3, AIM2); gasdermin D (GSDMD); cysteine-aspartic acid protease-1 (caspase-1); microRNA-153 (miR-153); nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB); inflammatory factors (IL-1β, IL-18). (b). Autophagy. After sensing external emergency signals, the cell activates AKT and AMPK or inhibits mTORC1, thereby activating the ULK1 complex and recruiting other autophagy-related proteins (ATG proteins) to initiate the formation of autophagosomes. The ATG5-ATG12-ATG16L1 complex is involved in membrane expansion, LC3-II is involved in autophagosome labeling, and mature autophagosomes fuse with lysosomes to form autolysosomes, degrading organelles and proteins. Excessive degradation or degradation disorders can lead to cell death. Autophagy-related genes (ATG); vacuolar protein sorting 34 (VPS34); Unc-51-like kinase 1 (ULK1); cathepsin B (CTSB); phosphatidylethanolamine (PE). (c). Cuproptosis. Cu+ inhibits Fe-S cluster protein synthesis and promotes protein lipid acylation. FDX1, ferredoxin 1. (d). Ferroptosis. Fe2+ produces ROS and thus causes ferroptosis. GPX4 inhibits ROS production and thus inhibits ferroptosis. TFR, transferrin receptor; STEAP3, six-transmembrane epithelial antigen of the prostate 3; DMT1, divalent metal transporter 1; ROS, reactive oxygen species; SLC7A11, solute carrier family 7 member 11; GSH, glutathione; GS-SH, oxidized glutathione disulfide; Nrf2, nuclear factor erythroid 2-related factor 2; GPX4, glutathione peroxidase 4. The diagram was drawn using Adobe Illustrator 2023.
Figure 3. Four cell death mechanisms in male infertility. (a). Pyroptosis. Some signaling molecules or stimuli activate pattern recognition receptors, thereby activating the inflammasome, activating caspase-1, cleaving the GSDMD protein, perforating the cell membrane, causing cell death, and releasing inflammatory factors to amplify the inflammatory response. Black arrows indicate activation or upregulation; adaptor protein (ASC); pattern recognition receptors (NLRP3, AIM2); gasdermin D (GSDMD); cysteine-aspartic acid protease-1 (caspase-1); microRNA-153 (miR-153); nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB); inflammatory factors (IL-1β, IL-18). (b). Autophagy. After sensing external emergency signals, the cell activates AKT and AMPK or inhibits mTORC1, thereby activating the ULK1 complex and recruiting other autophagy-related proteins (ATG proteins) to initiate the formation of autophagosomes. The ATG5-ATG12-ATG16L1 complex is involved in membrane expansion, LC3-II is involved in autophagosome labeling, and mature autophagosomes fuse with lysosomes to form autolysosomes, degrading organelles and proteins. Excessive degradation or degradation disorders can lead to cell death. Autophagy-related genes (ATG); vacuolar protein sorting 34 (VPS34); Unc-51-like kinase 1 (ULK1); cathepsin B (CTSB); phosphatidylethanolamine (PE). (c). Cuproptosis. Cu+ inhibits Fe-S cluster protein synthesis and promotes protein lipid acylation. FDX1, ferredoxin 1. (d). Ferroptosis. Fe2+ produces ROS and thus causes ferroptosis. GPX4 inhibits ROS production and thus inhibits ferroptosis. TFR, transferrin receptor; STEAP3, six-transmembrane epithelial antigen of the prostate 3; DMT1, divalent metal transporter 1; ROS, reactive oxygen species; SLC7A11, solute carrier family 7 member 11; GSH, glutathione; GS-SH, oxidized glutathione disulfide; Nrf2, nuclear factor erythroid 2-related factor 2; GPX4, glutathione peroxidase 4. The diagram was drawn using Adobe Illustrator 2023.
Ijms 26 03496 g003
Table 1. Different forms of cell death involved in male infertility caused by environmental pollutants.
Table 1. Different forms of cell death involved in male infertility caused by environmental pollutants.
Environmental PollutantExperimental Animal (Male)Experimental Cell TypeImpact on Male FertilityCell Death-Related PhenomenaForm of Cell DeathRef
Cadmium (Cd)Institute of Cancer Research (ICR) miceGC-1 spermatogonial cell line (GC-1)Cd exposure during puberty damaged testicular development and spermatogenesis in mice in adulthood.Cd caused iron overload, triggered ferroptosis in spermatogonia, and disturbed iron metabolism in spermatogonia.Ferroptosis[29]
C57BL/6 mice aged 6 weeks/Disruption of seminiferous tubules, extensive degenerative vacuolization, and intercellular gaps between germ cells.Elevated levels of LC3, p62, Atg7, Beclin-1, Atg5, and lysosomal membrane protein Lamp2.Autophagy[104]
Sprague-Dawley (SD) rats aged 10 weeksTM4 Sertoli cell line (TM4)BTB disruption with irregular and open Sertoli cell junctions.Accumulation of autophagic vacuoles and increased LC3-II levels.Autophagy[105]
CD-1 miceGC-1Enhanced testicular hyperemia and vacuolation.Elevated levels of N-GSDMD, cleaved caspase-1, and mature IL-1βPyroptosis[145]
Copper (Cu)ICR mice aged 8 weeksGC-1Reduction of spermatogonia and spermatocytes with vacuolar degeneration and necrosis of spermatogenic cells.Upregulation of Beclin-1, Atg5-Atg12, Atg7, Atg3, and Atg16L, and an increase in the LC3-II/LC3-I ratio.Autophagy[106]
Plasticizer di (2-ethylhexyl) phthalate (DEHP)ICR miceTM4Testicular atrophy decreased the organ coefficient of mouse testes and disrupted the BTB in Sertoli cells.DEHP induces ferroptosis in Sertoli cells, and its metabolite MEHP induces functional injury, disrupts glutathione metabolism disorder in TM4 cells, and accelerates ROS generation and lipid peroxidation.Ferroptosis[23]
Acrolein (ACR)/TM4/Increased Fe2+ levels, lipid peroxidation in Sertoli cells.Ferroptosis[33]
2,2′,4,4′-Tetrabromodiphenyl ether (PBDE-47)SD ratsTM4Reduced sperm count, increased the percentage of abnormal sperm, disrupted BTB integrity, and impaired testicular development.Upregulation of Fe2+ and MDA levels, and downregulation of GSH levels and Gpx4 and Slc7a11 protein levels in Sertoli cells and testes.Ferroptosis[27]
Cu sulfate (CuSO4)ICR mice/Dose-dependent testicular pathological disturbances, with reduced Sertoli cell and spermatogenic cell numbers.Elevated blood levels of Cu, Fdx1, and Slc7a11.Cuproptosis[73]
Cu oxide nanoparticles (CuONP)BALB/c mice/Testis development disorder and damaged sperm function and capacitation.Increased ROS and MDA levels, decreased GSH content.Cuproptosis[78]
Di-(2-ethylhexyl) phthalate (DEHP)SD ratsGC-1 and GC-2 spermatocyte cell line (GC-2)The testicular organ coefficient is low, the seminiferous epithelium is disordered, the number of germ cells is reduced, and the supporting cells are deformed.Upregulated PINK1 and PARKIN levels, and LC3 and COX IV co-localization.Autophagy[108]
Zinc oxide nanoparticles (ZnO NPs)/GC-1 Decreased vitality of GC-1 spg cells in mice.The protein content of LC3-II, the ratio of LC3-II/LC3-I, and the protein levels of ATG5 and Beclin-1 increased.Autophagy[107]
Tributyltin chloride (TBTCL)/Mouse Leydig cell line Reduced cell vitality.Decreased LC3-II and Beclin-1 levels, increased autophagic substrate p62 level.Autophagy[109]
Perfluorooctanoic acid (PFOA)BABL/c mice aged 8 weeks TM4Disruption of BTB integrity, reduction in sperm motility and count, seminiferous tubule damage, and significant decrease in seminiferous epithelium height.Upregulation of autophagy-related proteins LC3B and p62 in testicular Sertoli cells, and elevated levels of LC3-II/I, Beclin-1, and p62 in TM4 cells.Autophagy[110]
Zearalenone (ZEA)/Dairy goat Sertoli cellsInduction of oxidative stress and impairment of spermatogenesis.Increased the LC3-II/I ratio and decreased the level of p62 protein.Autophagy[102]
Aflatoxin B1 (AFB1)Kunming mice/Atrophy of seminiferous tubules, vacuolar changes in seminiferous epithelium, reduction in testicular interstitial cell and sperm count, significant increase in sperm deformity rate, significant decrease in sperm motility, and decrease in serum testosterone levels.Increased expression of LC3, Beclin-1, Atg5, Atg12, Atg13, and p62.Autophagy[103]
Cigarette/cigarette smoke condensate (CSC)Semen samplesGC-2Higher abnormalities of sperm viability and sperm progressive motility.Heavy smokers: Decreased GSH levels, increased lipid ROS and iron levels. CSC treatment effects: Decreased GSH level and GPX4 protein level, increased lipid ROS and iron levels in GC-2spd cells.Ferroptosis[21]
Monosodium glutamateWistar rats/Testicular degeneration, decreased testosterone, FSH, and LH levels, and abnormal sperm morphology.Elevated the levels of GSK-3β, NLRP3, caspase-1, and IL-1 β.Pyroptosis[146]
EthanolAdult Wistar rats/Inhibition of androgen receptor in testicular somatic cells.Elevated expression of LC3, accumulation of PINK1 and large lipid droplets.Autophagy[111]
PM2.5SD rats/Decreased the number of sperm in the testes, induced mitochondrial damage in testicular cells during sexual maturity in juvenile male rats, and disrupted iron metabolism. Decreased Gpx4 expression, reduced Slc7a11 expression.Ferroptosis[25]
/TM-3 cells, mouse Leydig cell lineDose-dependent decrease in TM-3 cell viability.Significant lipid peroxidation, elevated MDA levels, and increased ferrous content.Ferroptosis[22]
Fluoride (NaF) microplastics (MPs)BALB/c miceTM4Testicular injury, decreased Sertoli cell number, and functional impairment in mice.Gpx4 and downregulation in PS-MP and NaF + PS-MP groups.NaF and PS-MP co-exposure induces ferroptosis in Sertoli cells.Ferroptosis[28]
Microcystin and nitriteBalb/c miceGC-1 and TM4Decreased gonadal index and testosterone levels, reduced sperm density and survival rate, lowered number of spermatogonia.Increased caspase-1, N-GSDMD, and NLRP3 levels. Pyroptosis[142]
Silver nanoparticles (AgNPs)/GC-1 and GC-2/Increased NF–κB and NLRP3 protein levels.Pyroptosis[143]
FluorideC57BL/6J mice aged 6 weeks/Decreased sperm number and vitality, increased sperm abnormalities, reduced spermatogenic cell count, loosened testicular structure, and widened seminiferous tubule spacing.Upregulation of IL-18, IL-1β, LDH, NLRP3, AIM2, PYRIN, NLRP1, ASC, GSDMD, IL-1β, IL-18, Caspase-11, and Caspase-3.Pyroptosis[140]
1,2-DichloroethaneMiceGC-2 Testicular apoptosis, sperm malformation, and decreased sperm concentration.Elevated levels of NLRP3, GSDMD, N-GSDMD, Caspase-1, IL-1β, ASC, and IL-18, and formation of membrane pores in GC-2 spd cells.Pyroptosis[141]
Table 2. The potential mechanisms of key factors involved in various forms of cell death in male infertility.
Table 2. The potential mechanisms of key factors involved in various forms of cell death in male infertility.
Experimental AnimalExperimental Cell TypeForm of Cell DeathPhenomenaPossible MechanismRefs
/Fdx1 knockdown model in TM-3 cellsFerroptosisThe reduction in cell viability was alleviated, lipid peroxidation and cellular iron significantly declined after FDX1 knockdown.Upregulation of Fdx1 can promote ROS accumulation and ferrous overload in Leydig cells, induce lipid peroxidation, and lead to ferroptosis. After knocking down Fdx1, lipid peroxidation and cellular iron were significantly reduced, improving PM2.5-induced lipid peroxidation and ferrous accumulation.[22]
Nrf2−/− male mice/FerroptosisDecrease in fertility, decreases in sperm concentration and motility, inhibition of ferroptosis attenuated spermatogenic cell damage in the testes and epididymis, and increases in sperm concentration and motility.Gpx4 and Slc7a11 significantly decreased.[59]
S8/Gss/ male mice/FerroptosisThere was a decline in Gpx4 and an increase in Alox15 levels observed in 8-month-old S8/Gss−/− mice, resulting in the accumulation of ROS, lipid peroxidation, and ultimately testicular ferroptosis.Gss deficiency caused ferroptosis in the testes of mice, testicular ferroptosis may cause meiosis disruption and acrosome heterotopia. The resulting aberrant sperm showed lower concentrations and abnormal morphology, leading to reduced fertility.[54]
Atg5flox/flox; Stra8-iCre/Autophagy70% of mice are infertile, with reduced sperm count and motility and impaired sperm morphology.The absence of ATG5 significantly reduces the expression of testicular LC3A/B-II, increases the expression of autophagy receptor SQSTM1/p62, and reduces testicular autophagy activity.[127]
Becn1flox/Δ; Stra8-Cre/AutophagyThe fertility of mice decreased, with a significant decrease in sperm motility and count, as well as abnormalities in sperm morphology and structure.The absence of Beclin-1 leads to the accumulation of p62, a decrease in Atg5, and inhibition of the autophagic process.[128]
Gsdmdfl/flCx3cr1-cre/PyroptosisImproved sperm quality and reduced the expression of pro-inflammatory cytokines IL-1 β and TNF-α.Specific knockout of GSDMD in macrophages can reduce the expression of pro-inflammatory cytokines IL-1 β and TNF-α and improve testicular injury induced by orchitis.[153]
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

Cai, D.; Li, J.; Peng, Z.; Fu, R.; Chen, C.; Liu, F.; Li, Y.; Su, Y.; Li, C.; Chen, W. Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities. Int. J. Mol. Sci. 2025, 26, 3496. https://doi.org/10.3390/ijms26083496

AMA Style

Cai D, Li J, Peng Z, Fu R, Chen C, Liu F, Li Y, Su Y, Li C, Chen W. Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities. International Journal of Molecular Sciences. 2025; 26(8):3496. https://doi.org/10.3390/ijms26083496

Chicago/Turabian Style

Cai, Difan, Junda Li, Zekang Peng, Rong Fu, Chuyang Chen, Feihong Liu, Yiwang Li, Yanjing Su, Chunyun Li, and Wei Chen. 2025. "Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities" International Journal of Molecular Sciences 26, no. 8: 3496. https://doi.org/10.3390/ijms26083496

APA Style

Cai, D., Li, J., Peng, Z., Fu, R., Chen, C., Liu, F., Li, Y., Su, Y., Li, C., & Chen, W. (2025). Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities. International Journal of Molecular Sciences, 26(8), 3496. https://doi.org/10.3390/ijms26083496

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