Impact of Heat-Stress-Induced Oxidative Stress and Apoptosis: Comparison
Please note this is a comparison between Version 2 by Mona Zou and Version 1 by MUHAMMAD ZAHOOR KHAN.

Heat stress represents a pervasive global concern with far-reaching implications for the reproductive efficiency of both animal and human populations. An extensive body of published research on heat stress effects utilizes controlled experimental environments to expose cells and tissues to heat stress and its disruptive influence on the physiological aspects of reproductive phenotypic traits, encompassing parameters such as sperm quality, sperm motility, viability, and overall competence. Beyond these immediate effects, heat stress has been linked to embryo losses, compromised oocyte development, and even infertility across diverse species. One of the primary mechanisms underlying these adverse reproductive outcomes is the elevation of reactive oxygen species (ROS) levels precipitating oxidative stress and apoptosis within mammalian reproductive cells. Oxidative stress and apoptosis are recognized as pivotal biological factors through which heat stress exerts its disruptive impact on both male and female reproductive cells. In a concerted effort to mitigate the detrimental consequences of heat stress, supplementation with antioxidants, both in natural and synthetic forms, has been explored as a potential intervention strategy. Furthermore, reproductive cells possess inherent self-protective mechanisms that come into play during episodes of heat stress, aiding in their survival. 

  • heat stress
  • oxidative stress
  • apoptosis
  • antioxidants
  • mammalian reproductive cells
  • fertility

1. Introduction

Heat stress refers to physiological responses occurring due to prolonged exposures to high temperatures, often combined with elevated humidity, that cause body heat gain to exceed one’s ability to dissipate the heat [1]. It arises when the body is unable to thermoregulate properly, leading to an abnormal rise in core body temperature [2]. The fundamental origin of escalating heat stress is global warming stemming from anthropogenic climate change—the accumulation of greenhouse gases like carbon dioxide, methane, and nitrous oxides due to human activities including burning fossil fuels, deforestation, and agriculture [3]. One key mechanism is through the impairment of evaporative cooling from sweating and respiration [4]. High humidity in particular hampers heat loss via sweating and skin evaporation by reducing the vapor pressure gradient from the skin to the environment [5]. Studies show that humid heatwaves have become more frequent worldwide [6], causing instances of near-fatal and fatal hyperthermia and heat stroke by blocking the primary heat loss avenue in humans [7]. The multifaceted effects of escalating heat stress can be detrimental with widespread repercussions for health, economies, ecosystems, and more. Some major anticipated impacts include dramatic rises in heat-related compromised reproductive efficiency [8]. The deleterious effects of heat stress (HS) on reproductive function encompass a comprehensive spectrum of repercussions affecting both male and female reproductive components, spanning from the intricate mechanisms of fertilization to the critical stages of early and late embryo–fetal development. Consistently, extensive investigations have collectively unveiled the multifaceted impacts of HS on the reproductive landscape [9,10,11,12,13][9][10][11][12][13].
Within the sphere of male reproductive physiology, HS emerges as a pivotal factor intricately linked to male infertility, exerting a discernible influence on testicular functionality [13,14][13][14]. Notably, empirical evidence drawn from diverse studies underscores the detrimental ramifications of HS on male fertility and semen quality [15,16,17,18,19,20,21,22][15][16][17][18][19][20][21][22]. Furthermore, HS casts a shadow over the ensuing fertilization processes, as elucidated by recent investigations [23,24,25][23][24][25]. Recent scientific inquiries have accentuated the detrimental influence of HS on sperm quality across a spectrum of mammalian species, encompassing dogs [17], bulls [20], buffaloes [26], stallions [27[27][28][29],28,29], rabbits [30], pigs [31], goats [32], and human males [33,34][33][34]. Remarkably, a recurring theme in these studies underscores the adverse impact of HS-induced oxidative stress and apoptosis on the viability of Sertoli cells, spermatogonial stem cells, Leyding cells, spermatogenesis, and sperm quality [15,35,36,37,38,39,40,41,42][15][35][36][37][38][39][40][41][42]. Furthermore, recent publications have elucidated a compelling association between sperm quality and environmental temperature in men, underscoring the pervasive implications of environmental factors on male reproductive health [43,44,45,46][43][44][45][46]. These findings collectively reinforce the imperative need for comprehensive research into mitigating the detrimental effects of HS on reproductive function across species, as well as the exploration of potential interventions to safeguard male fertility and sperm quality under challenging environmental conditions.
In the United States, cattle reproduction and milk production rates experience a pronounced decline during the hot season due to HS, resulting in approximately USD 900 million in annual losses within the dairy industry [47,48][47][48]. Besides the metabolic stress associated with high milk production in dairy cows, seasonal impacts on fertility have been extensively documented [49,50][49][50]. Notably, a substantial reduction in cattle fertility during the summer months has been attributed to the thermoregulatory challenges posed by HS, leading to elevated body temperatures [51,52,53][51][52][53]. Furthermore, HS exerts detrimental effects on follicle quality and hormonal equilibrium, contributing to a decline in estrus [54,55,56,57,58][54][55][56][57][58]. Specifically, granulosa cells responsible for estradiol production are adversely affected by HS, resulting in decreased estradiol production and subsequent disruption of the estrous cycle in cattle [59,60,61][59][60][61]. HS has also been shown to influence corticoid levels, luteinizing hormone (LH), and plasma progesterone in cattle, with alterations in hypothalamus and pituitary gland function leading to changes in the secretion of reproductive hormones [9,62,63,64,65,66][9][62][63][64][65][66]. Moreover, HS negatively impacts oocyte quality due to its effects on the hypothalamus and pituitary gland, specifically the LH [67,68,69][67][68][69]. Additionally, Khan et al. [47] reported that HS significantly compromises granulosa cells, leading to suboptimal oocyte development. This is further supported by studies that found HS compromises the mRNA expression levels of Moloney sarcoma oncogene (MOS), growth factor 9 (GDF9), and POU domain, class 5, transcription factor 1 (POUF51), which are crucial for oocyte development and competence, ultimately resulting in poor-quality oocytes and impaired embryonic development [60,61,70,71,72][60][61][70][71][72]. Figure 1 demonstrates the adverse impact of heat stress on phenotypic traits related to mammalian reproductive traits.
Figure 1. Effect of antioxidant supplementation on mammalian reproductive cells under heat stress: pre- and post-supplementation comparison.
In response to these challenges, numerous researchers have advocated for exogenous supplementation with antioxidants to mitigate the elevated levels of reactive oxygen species (ROS) induced by HS and enhance the antioxidant capacity of mammalian reproductive cells [30,31,47,73,74,75,76][30][31][47][73][74][75][76]. Consistently, positive outcomes have been observed for supplementation with antioxidants, whether in the form of herbal medicines or feed additives, in improving the efficiency of mammalian reproductive cells such as granulosa cells, Leydig cells, and Sertoli cells by alleviating oxidative stress [77,78,79,80,81,82,83,84,85][77][78][79][80][81][82][83][84][85]. The beneficial impact of antioxidant supplementation in mitigating the adverse effects of heat stress on phenotypic traits related to mammalian reproduction has been succinctly illustrated in Figure 1

2. Impact of Heat-Stress-Induced Oxidative Stress and Apoptosis on Mammalian Reproductive Cell Functionality

It is well established that heat stress induces ROS production primarily through mitochondrial dysfunction, where the electron transport chain is compromised, leading to electron leakage and the formation of superoxide radicals (O−2) [86,87][86][87]. Additionally, another crucial source of ROS induction during heat stress is the activation of NADPH oxidases (NOXs), particularly NOX2 and NOX4. These enzymes are responsible for generating superoxide ions in response to various stressors, including heat. The activation of NOX enzymes can occur through heat-induced signaling pathways, such as the activation of protein kinase C (PKC) and mitogen-activated protein kinases (MAPKs). The NADPH oxidases also contribute to the production of superoxide radicals during heat stress [88]. Moreover, these superoxide radicals can be further converted into other types of ROS, such as hydrogen peroxide (H2O2) and hydroxyl radicals (OH), through various reactions, including those catalyzed by superoxide dismutase (SOD) [89,90][89][90]. Consequently, heat-stress-induced ROS can also trigger lipid peroxidation, leading to the formation of lipid peroxidation products such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE). These products are known to be cytotoxic and can disrupt cellular membranes [90,91][90][91]. These molecules, while playing roles in signaling under normal physiological conditions, can cause significant damage to cellular structures and DNA when present in excess due to heat stress [90,91,92][90][91][92].
Oxidative damage and cell apoptosis represent pivotal consequences affecting mammalian reproductive cells, primarily initiated by the excessive generation of ROS [93,94,95][93][94][95]. The accrual of ROS disrupts male reproductive functions and exerts detrimental effects on semen quality [96,97][96][97]. These detrimental effects manifest in two main modes: firstly, the overabundance of ROS depletes the cell’s scavenging capacity, impairs antioxidant enzymes, escalates lipid peroxidation, and triggers DNA damage, ultimately compromising the cell’s defense against oxidative harm. Secondly, the surplus ROS mediate molecular signaling in the mitochondria-dependent apoptotic pathway, encompassing events such as the opening of mitochondrial permeability transition pores, mitochondrial membrane depolarization, and the release of mitochondrial substances, including cytochrome C (cyto-c). This, in turn, culminates in caspase-3 activation and subsequent cell apoptosis. Recent research by Li H et al. [98] further reported that heat stress upregulated caspase-3 and caspase-9, leading to enhanced apoptosis in endometrial epithelial and glandular epithelial cells, along with alterations in HO-1 mRNA/protein and Keap1 mRNA/protein expression, and an elevated malondialdehyde (MDA) level in mouse uterine tissue.
In the context of male reproductive cells, recent investigations have extensively explored the impact of heat stress on testis morphology, antioxidant status, and testicular biosynthesis [34,99,100,101][34][99][100][101]. Sertoli cells, known for providing structural and nutritional support for developing germ cells, have been a focus of scrutiny, with studies comprehensively examining the repercussions of heat stress on male reproductive cells, including Sertoli cells, spermatogonial stem cells, and Leydig cells [102]. Wang C et al. [103] reported that HS induces oxidative stress and apoptosis in Sertoli cells, disrupting the normal spermatogenesis process. Furthermore, boar Sertoli cells exposed to elevated temperatures exhibited increased oxidative stress and apoptosis, an inhibited pentose phosphate pathway, and decreased ATP content. Molecular changes observed in boar Sertoli cells under heat stress involved the downregulation of the Kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway (associated with enhanced antioxidants) and low levels of heat shock protein 90 (HSP90) due to the suppression of melatonin receptor 1B (MTNR1B), resulting in abnormal regulation of stabilizing hypoxia-inducible factor-1α (HIF-1α) [104]. Another study by Xue H et al. [105] found that HS primarily enhanced the lipid oxidation, oxidative stress, and apoptosis in Sertoli cells through the activation of arachidonate 15-lipoxygenase type B (ALOX15B) and the production of 8-hydroxyeicosatetraenoic acid (8-HETE) and 15-hydroxyeicosatetraenoic acid (15-HETE), with involvement of the P53-p38 pathway [105]. The disruption of arachidonic acid (AA) metabolism, a precursor to 20-carbon polyunsaturated fats, has been reported to be associated with poor spermatogenesis outcomes, as excessive AA levels altered cytomembrane structure and function and increased permeability and brittleness, potentially leading to mitochondrial changes, apoptosis, or necrosis. HS was found to significantly elevate AA levels, disrupting the function of tight junctions (TJs) essential for spermatogenesis development [106]. Additionally, AA increased MDA levels, activated p38 mitogen-activated protein kinases (P38 MAPKs), and reduced mitochondrial DNA (mtDNA). Furthermore, another study noted that heat stress induced oxidative stress in Sertoli cells by suppressing the level of nuclear factor erythroid 2-related factor 2 (Nrf2) [107]. In addition, the effects of oxidative stress on Leydig cells have been briefly reviewed in recent studies [102,108][102][108]. Heat stress treatment inhibited cell viability, induced apoptosis, increased the activity of caspase 3 and the pro-apoptotic protein Bax, and decreased the expression of anti-apoptotic protein B-cell leukemia/lymphoma-2 (Bcl-2), concurrently activating endoplasmic reticulum (ER) stress markers such as glucose-regulated protein 78 (GRP78) and CCAAT/enhancer-binding protein homologous protein (CHOP) [109].
The impact of heat-stress-induced oxidative stress and apoptosis on mammalian female reproductive cells has also been extensively documented in recent research [110]. In alignment with these findings, it has been observed that heat-stress-induced oxidative stress and apoptosis in bovine granulosa cells disrupt the normal secretion of estrogen, leading to disturbances in the ovarian microenvironment and subsequent interference with ovarian function [111]. Consistently, a study documented abnormal folliculogenesis including impaired ovulation, fertilization, and early embryo development [112]. Additionally, a study has reported elevated levels of ROS production in response to heat stress, resulting in increased apoptosis and even embryo death, coupled with reductions in both mitochondrial activity and membrane potential [113]. Consistently, Sammad et al. [114] have reported elevated levels of ROS and apoptosis in bovine granulosa cells under heat stress conditions. They also noted the negative regulation of several candidate genes, including heme oxygenase 1 (HMOX1), nitric oxide synthase 2 (NOS2), catalase (CAT), superoxide dismutase (SOD), B-cell lymphoma 2-like 1 (BCL2L1), glutathione peroxidase 4 (GPX4), Nrf2, aspartoacylase 3 (ASP3), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGCIA), solute carrier family 16 member 3 (SLC16A3), sterol regulatory element-binding protein 1 (SERBP1), sirtuin 1 (SIRT1), AMP-activated protein kinase (AMPK), Caspase 8 (CASP8), CASP9, insulin-like growth factor 2 (IGF2), peroxisome proliferator-activated receptor alpha (PPARA), and solute carrier family 27 member 3 (SLC27A3), which are associated with apoptosis, cell proliferation, and oxidative activity of granulosa cells [114]. Furthermore, their research indicated that heat stress significantly downregulated the key anti-apoptotic and antioxidant-associated signaling pathways, including the AMP and Nrf2 signaling pathways [114]. Similarly, another study reported a significant decrease in the number of primordial follicles, an increase in the number of degenerated follicles, and a decrease in granulosa cell proliferation in response to heat stress [115]. The molecular mechanisms associated with heat-stress-induced oxidative stress and apoptosis effects on mammalian reproductive cells are summarized in Figure 2 and Table 1.
Figure 2. Cellular and molecular responses to heat-stress-induced oxidative stress and cell apoptosis in mammalian reproductive cells. The “→” shows direct correlation/effect, while “Antioxidants 13 00258 i001” indicates the effect has been suppressed.
Table 1. Effect of heat stress on mammalian reproductive cells.
Heat Stress Biological Effect Cells Reference
 
Enhanced accumulation of ROS, suppressed SOD, CAT, and proliferating cell nuclear antigen (PCNA) protein expression levels
Sertoli cells [116]
 
Elevates the level of arachidonic acid which disrupts TJs and enhances oxidative stress and apoptosis
Disrupts the normal process of spermatogenesis
Sertoli cells [106]
 
Elevated the transforming growth factor beta 1 (TGFβ1)/SMAD family member (Smad2)/Smad3 pathway protein expression, causing cell apoptosis, testicular tissue organic lesions, and testicular damage and affecting testicular secretion function.
Testis [117]
 
Increased the process of apoptosis by enhancing the level of Bcl-2-associated X protein (BAX) and Caspase-3
Suppressed cell proliferation by downregulating PCNA and CyclinB1
Enhanced ROS and oxidative stress by suppressing the level of SOD
Disrupted the synthesis of progesterone and estrogen by downregulation of the expression of steroidogenic acute regulatory protein (STAR), Cyp11A1
Ovarian granulosa cells [95]
 
Induced apoptosis via endoplasmic reticulum stress signaling
Upregulates the expression of apoptotic linked genes (caspase-3, BAX, glucose-regulated protein 78 (GRP78) and CHOP) and downregulates BCL2 gene expression
Mouse granulosa cells [118]

3. Advancement and Understanding of Genetic Biomarkers Associated with Heat Stress Resistance and Reduced Apoptosis and Oxidative Stress in Mammalian Reproductive Cells

It is well established that heat resistance in mammalian reproductive cells is mediated by a network of genes and their signaling pathways to counteract the damaging effects of heat stress. Based on published data, several genes and pathways, including HSP family genes; anti-apoptotic genes; genes encoding antioxidant enzymes; and the AMPK, ERK1/2, and Nrf2/Keap1 signaling pathways, have been found to be involved in protective mechanisms against heat-stress-induced apoptosis and oxidative stress in mammalian reproductive cells. Detailed information regarding the protective role of the aforementioned genes and pathways against heat stress in mammalian reproductive cells is provided below.

3.1. Role of Heat Shock Protein (HSP) Genes in Mitigating Heat-Stress-Induced Oxidative Stress and Apoptosis in Mammalian Reproductive Cells

The heat shock protein-72 (Hsp72) gene, a prominent member of the heat shock protein (HSP) family, serves as the primary inducible heat shock protein. Its baseline expression in healthy cells is minimal, but it becomes markedly upregulated in response to heat stress. Notably, HSP72 has demonstrated its ability to counteract heat-stress-induced ROS in bovine Sertoli cells when exposed to puerarin, a traditional Chinese medicinal compound [13]. In this context, HSP72 functions as both an antioxidant and an anti-apoptotic factor within Sertoli cells. It achieves this by reducing ROS production and safeguarding Sertoli cells against oxidative harm and apoptotic processes. Consistently, a study reported the protecting and self-recovering role of HSP70 in bovine oocytes after exposure to severe heat stress [119]. Furthermore, it revealed that HSP70 prevents apoptosis, supports signal transduction, increases the antioxidant protection of the embryo, as well as protecting heat-stressed maturing bovine oocytes and restoring their developmental competence. Consistently, Ho et al. [59] observed that Asparagus officinalis stem (EAS) elicited an upregulation of HSP70 and heat shock factor 1 (HSF1) expression, resulting in an augmented concentration of progesterone within heat-treated bovine cumulus–granulosa cells. Additionally, EAS demonstrated the capacity to heighten glutathione (GSH) levels, improve mitochondrial function, and mitigate ROS levels in heat-stressed bovine cumulus–granulosa cells. Notably, when HSP70 was inhibited by Ho et al., a subsequent decrease was noted in the levels of progesterone, GSH, HSF1, Nrf2, and Kelch-like ECH-associated protein 1 (Keap1). These findings collectively underscore the pivotal role of HSP70 as the principal regulator orchestrating antioxidant activity, thereby safeguarding granulosa cells against the deleterious effects of heat stress [59].
Heme oxygenase 1 (HO-1), alternatively known as heat shock protein-32 (Hsp32), is a stress-responsive enzyme with pivotal roles in maintaining iron homeostasis, fortifying antioxidant defenses, and averting apoptosis [120,121,122][120][121][122]. Interestingly, studies have disclosed an association between low serum levels of HO-1 and an elevated risk of polycystic ovarian syndrome [123]. Furthermore, investigations have substantiated that HO-1 attenuates heat-stress-induced apoptosis in bovine granulosa cells by curbing ROS production and activating antioxidant responses [124]. Remarkably, HO-1 modulation influences apoptotic processes, with its downregulation intensifying apoptosis and its upregulation mitigating apoptosis through the regulation of Bax/Bcl-2 expression and cleaved caspase-3 levels [111]. Additionally, HO-1 plays a cytoprotective role by influencing estrogen levels and catalyzing the breakdown of heme to generate biologically active carbon monoxide (CO). Significantly, CO elevation coincides with heightened HO-1 levels, diminished Bax/Bcl-2 ratios, and inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway (Figure 2) [111]. Studies have shown that reducing antioxidant gene levels in heat-stressed (40 °C) HO-1-knockdown bovine granulosa cells leads to increased cellular apoptosis [124]. Moreover, research has elucidated that heat stress triggers the activation of Nrf2, which safeguards bovine granulosa cells from heat-stress-induced apoptosis by regulating HO-1. This, in turn, modulates ROS levels, reducing their production and subsequently suppressing oxidative stress and apoptosis [124].

3.2. Protective Role of SOD Genes against Heat-Stress-Induced Oxidative Stress and Apoptosis in Mammalian Reproductive Cells

The SOD genes’ protective role against heat stress has been established in mammalian reproductive cells [95,125][95][125]. Khan et al. conducted experimental studies demonstrating that the silencing of the SOD1 gene in heat-treated granulosa cells resulted in increased apoptosis, reduced cell proliferation, and decreased biosynthesis of estrogen and progesterone hormones, as depicted in the accompanying Figure 3 [95]. In addition, Faheem et al. conducted a study where they observed that under conditions of heat stress, buffalo granulosa cells demonstrated elevated expression levels of SOD2 and an enhancement in total antioxidant activity [126]. Furthermore, Faheem et al. highlighted the enhanced antioxidant capacity and cholesterol levels in granulosa cells, which likely contribute significantly to their biological function in preventing heat-stress-induced apoptosis and oxidative stress [125]. These findings suggest that SOD1 plays a key role in regulating other genes while protecting buffalo granulosa cells from the adverse effects of heat stress.
Figure 3. Silencing of SOD1 genes may lead to elevated levels of ROS followed by disruption of steroidogenesis, compromised cell proliferation, and increased cell apoptosis. The “→” shows direct correlation/effect, while “Antioxidants 13 00258 i001” indicates the effect has been suppressed.

3.3. ERK1/2 Signaling Pathway Protects Mammalian Reproductive Cells from Heat-Stress-Induced Apoptosis

The ERK1/2 kinases are highly conserved serine–threonine kinases with widespread distribution, playing a pivotal role in cellular signaling regulation, both in normal physiological conditions and pathological states, by phosphorylating various substrates. In response to heat stress, ERK1/2 initiates a series of cascading reactions that modulate the balance between cellular survival and apoptosis molecules, thereby safeguarding a portion of male germ cells and somatic cells within the testis from destruction. Research indicates that a brief 30 min exposure to heat stress triggers an increase in phosphorylated ERK1/2 (pERK1/2) levels in immature boar Sertoli cells. This, in turn, elevates HSP70 levels and subsequently enhances the production of lactate, a primary ATP substrate crucial for the development of germ cells, by accelerating glucose metabolism [127]. Furthermore, ERK signaling exerts protective effects on pachytene spermatocytes subjected to transient heat stress by upregulating metastasis-associated 1, which counteracts the pro-apoptotic effects of p53 [128]. Conversely, inhibiting the ERK1/2 signaling pathway during heat stress significantly reduces the expression of genes such as c-fos, AP-1, and ERK2, as well as the phosphorylation of ERK1/2 and c-Fos. This inhibition is accompanied by a marked increase in c-Jun mRNA expression within Sertoli cells. Notably, the adverse effects of heat stress on the ERK1/2 signaling pathway can be ameliorated through treatment with baicalin [113]. Consistently, Wang et al. have reported that heme oxygenase 1 (HO-1) utilizes the ERK1/2 signaling pathway to suppress the expression of apoptotic genes, specifically Bax/Bcl-2, thereby restoring the normal functionality of bovine granulosa cells [111]. This interplay between ERK1/2 signaling and HO-1 underscores their pivotal roles in modulating cellular responses to heat stress, ultimately influencing cell survival and apoptosis in reproductive cells.

3.4. Protective Role of Nrf2 in Protection of Mammalian Cells against Heat-Stress-Induced Oxidative Stress and Apoptosis

Nrf2 is an inducible transcription factor crucial for maintaining redox signaling integrity in the face of oxidative stress [107]. Nrf2, a member of the Cap’n’Collar basic leucine zipper transcription factor family, plays a pivotal role in orchestrating antioxidant and detoxification responses through the upregulation of its downstream genes [129]. In unstressed cells, the Nrf2 is primarily located within the cellular cytoplasm and forms a complex with its inhibitory partner, Kelch-like ECH-associated protein 1 (Keap1). However, when the cellular environment encounters an elevated presence of ROS, the Keap1-Nrf2 complex undergoes dissociation, leading to the translocation of Nrf2 from the cytoplasm into the cellular nucleus [130,131][130][131]. Furthermore, the activated Nrf2 binds to the antioxidant response element (ARE) sequence, thereby stimulating the transcription of genes involved in antioxidant defenses and neutralizing ROS-induced damage [115]. Recent findings indicate that p62 can competitively interact with Keap1 at the Nrf2 binding site, altering the association, releasing ubiquitinated Nrf2, and ultimately activating the Nrf2 antioxidant systems [132,133,134][132][133][134].
Heat-stress-induced apoptosis triggers antioxidant responses, including autophagy and Nrf2 activation [135]. It has been observed that changes in autophagy dynamics are pivotal regulators of the Nrf2 signaling pathway’s protective role in the testis. This protection is achieved by suppressing MDA levels and promoting an antioxidant status that shields the testis from the adverse consequences of heat stress [136,137,138,139][136][137][138][139]. Notably, inhibiting Nrf2 in cells results in reduced cell viability, elevated MDA levels, and Sertoli cell death [107].
Nrf2 is known to regulate several crucial antioxidant genes, including catalase (CAT), heme oxygenase 1 (HMOX1), peroxiredoxin 1 (PRDX1), SOD1, and thioredoxin 1 (TXN1). These genes collectively enhance antioxidant activity, mitigating oxidative stress in mouse testis cells and protecting germ cells and Leydig cells from oxidative damage [56,139][56][139]. Furthermore, recent research has shown that heat-stress-induced ROS overproduction suppresses the expression of antioxidant genes (SOD, CAT, NQO1, and GSH-Px) in uterine tissue [98]. In Sertoli cells, elevated ROS levels due to heat stress increase MDA levels and decrease antioxidant enzyme levels [140]. Additionally, heat stress has been found to increase the expression of apoptotic markers such as Fas, FasL, caspase 3, and caspase 9 in mouse Sertoli cells [140]. Consequently, the Keap1/Nrf2 signaling pathway has been significantly associated with the protective effects observed in mouse uterine tissue, marked by increased levels of antioxidant genes [98].
Moreover, oxidative stress influences various important signaling pathways, including the nuclear factor erythroid 2-related factor 2 (Nrf2)/Keap1 signaling axis in the testis [141]. A recent study highlights Nrf2’s protective role in safeguarding mouse Sertoli cells from heat-induced oxidative stress through the Nrf2/Keap1 signaling pathway [107]. Similarly, another investigation revealed that Nrf2 significantly reduces caspase 3 levels, subsequently reducing cell death induced by heat stress treatment in Sertoli cells [137]. Under conditions of severe heat stress, the heightened expression of Keap1 and NFE2L2 facilitates the regulation of genes associated with antioxidants by forming complexes with the ARE, thus establishing a defensive mechanism against heat stress within bovine endometrial epithelial cells [142]. These findings collectively underscore the critical role of Nrf2 in mitigating oxidative stress and apoptosis in various cellular contexts, particularly under heat stress conditions.

3.5. Role of Adenosine 5′-Monophosphate-Activated Protein Kinase (AMPK) in Self-Recovery from Heat-Stress-Induced Oxidative Stress and Apoptosis

It is well established that AMPK signaling plays a key role in the tight junctions (TJs) and cell proliferation of testis Sertoli cells [143,144,145,146,147,148][143][144][145][146][147][148]. In addition, Ni et al. [147] highlighted that Sertoli cells play a key role in lactate supply, maintenance of cell junctions, and support for germ cells’ mitosis and meiosis. The AMPK signaling pathway regulates the dynamics of tight junctions and adherens junctions; the proliferation and meiosis of germ cells; and the energy metabolism, proliferation, and lactate production of Sertoli cells [149]. Once this balance is disrupted, the microenvironment of the testis and the quality of sperm will be affected. When α1AMPK was conditionally knocked out in mouse SCs, the mutant mice still showed an abnormal phenotype, including thin-head spermatozoa, reduced expression of junctional proteins (β-catenin, vimentin, occludin, and ZO-1), and deregulation of energy homeostasis [143,145,147,148][143][145][147][148]. Consistently, a study has documented that curcumin (natural antioxidant and anti-inflammatory compound) supplementation rescues porcine Sertoli cell impairment and TJs by inhibiting the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome through the AMPK/SIRT3/SOD2/mtROS signaling pathway [150]. Heat stress can cause dysfunction of TJs in porcine testis reversibly via Ca2+/calmodulin-dependent protein kinase kinase B (CaMKKB)-induced inhibition of the AMPK signaling pathway. Consistently, Yang et al. [148] treated SCs from 3-week-old piglets at 43 °C for 0.5 h, and this hyperthermia treatment inhibited the AMPK signaling pathway, inhibiting the expression of CLDN11, JAMA, occludin, and especially ZO-1 in porcine SCs [148]. In addition, it was observed that normal Sertoli cell function was restored after 48 h due to AMP signaling [145].

References

  1. Becker, C.A.; Collier, R.J.; Stone, A.E. Invited review: Physiological and behavioral effects of heat stress in dairy cows. J. Dairy Sci. 2020, 103, 6751–6770.
  2. Schulte, P.A.; Bhattacharya, A.; Butler, C.R.; Chun, H.K.; Jacklitsch, B.; Jacobs, T.; Kiefer, M.; Lincoln, J.; Pendergrass, S.; Shire, J.; et al. Advancing the framework for considering the effects of climate change on worker safety and health. J. Occup. Environ. Hyg. 2016, 13, 847–865.
  3. Ruane, A.C.; Vautard, R.; Ranasinghe, R.; Sillmann, J.; Coppola, E.; Arnell, N.; Cruz, F.A.; Dessai, S.; Iles, C.E.; Islam, A.K.M.S.; et al. The Climatic Impact-Driver Framework for Assessment of Risk-Relevant Climate Information. Earths Future 2022, 10, e2022EF002803.
  4. Shibasaki, M.; Crandall, C.G. Mechanisms and controllers of eccrine sweating in humans. Front. Biosci. 2010, 2, 685–696.
  5. Hanna, E.G.; Tait, P.W. Limitations to Thermoregulation and Acclimatization Challenge Human Adaptation to Global Warming. Int. J. Environ. Res. Public Health 2015, 12, 8034–8074.
  6. Thompson, V.; Mitchell, D.; Hegerl, G.C.; Collins, M.; Leach, N.J.; Slingo, J.M. The most at-risk regions in the world for high-impact heatwaves. Nat. Commun. 2023, 14, 2152.
  7. Walter, E.J.; Carraretto, M. The neurological and cognitive consequences of hyperthermia. Crit. Care 2016, 20, 199.
  8. Zhang, G.; Han, L.; Yao, J.; Yang, J.; Xu, Z.; Cai, X.; Huang, J.; Pei, L. Assessing future heat stress across China: Combined effects of heat and relative humidity on mortality. Front. Public Health 2023, 11, 1282497.
  9. Dovolou, E.; Giannoulis, T.; Nanas, I.; Amiridis, G.S. Heat Stress: A Serious Disruptor of the Reproductive Physiology of Dairy Cows. Animals 2023, 13, 1846.
  10. Tirpák, F.; Halo, M.; Tomka, M.; Slanina, T.; Tokárová, K.; Błaszczyk-Altman, M.; Dianová, L.; Ivanič, P.; Kirchner, R.; Greń, A.; et al. Sperm Quality Affected by Naturally Occurring Chemical Elements in Bull Seminal Plasma. Antioxidants 2022, 11, 1796.
  11. Tirpák, F.; Greifová, H.; Lukáč, N.; Stawarz, R.; Massányi, P. Exogenous factors affecting the functional integrity of male reproduction. Life 2021, 11, 213.
  12. Boni, R. Heat stress, a serious threat to reproductive function in animals and humans. Mol. Reprod. Dev. 2019, 86, 1307–1323.
  13. Cong, X.; Zhang, Q.; Li, H.; Jiang, Z.; Cao, R.; Gao, S.; Tian, W. Puerarin ameliorates heat stress–induced oxidative damage and apoptosis in bovine Sertoli cells by suppressing ROS production and upregulating Hsp72 expression. Theriogenology 2017, 88, 215–227.
  14. Cao, K.X.; Deng, Z.C.; Liu, M.; Huang, Y.X.; Yang, J.C.; Sun, L.H. Heat Stress Impairs Male Reproductive System with Potential Disruption of Retinol Metabolism and Microbial Balance in the Testis of Mice. J. Nutr. 2023, 153, 3373–3381.
  15. Costa, G.M.; Lacerda, S.M.; Figueiredo, A.F.; Leal, M.C.; Rezende-Neto, J.V.; França, L.R. Higher Environmental Temperatures Promote Acceleration of Spermatogenesis in Vivo in Mice (Mus musculus). J. Therm. Biol. 2018, 77, 14–23.
  16. Huang, D.; Cai, J.; Zhang, C.; Jin, R.; Bai, S.; Yao, F.; Ding, H.; Zhao, B.; Chen, Y.; Wu, X.; et al. Semen quality and seminal plasma metabolites in male rabbits (Oryctolagus cuniculus) under heat stress. PeerJ 2023, 11, e15112.
  17. Lyrio, L.L.; Lazaro, M.A.; Sonegheti, R.; Moulin, L.; Coslop, L.; Sarto, C.G.; Loureiro, B.; Favoreto, M.G. Effects of heat stress on sperm quality of French Bulldogs. Theriogenology 2023, 199, 131–137.
  18. Takalani, N.B.; Monageng, E.M.; Mohlala, K.; Monsees, T.K.; Henkel, R.; Opuwari, C.S. Role of oxidative stress in male infertility. Reprod. Fertil. 2023, 4, 3.
  19. Netherton, J.K.; Robinson, B.R.; Ogle, R.A.; Gunn, A.; Villaverde, A.I.; Colyvas, K.; Wise, C.; Russo, T.; Dowdell, A.; Baker, M.A. Seasonal variation in bull semen quality demonstrates there are heat-sensitive and heat-tolerant bulls. Sci. Rep. 2022, 12, 15322.
  20. Morrell, J.M. Heat stress and bull fertility. Theriogenology 2020, 153, 62–67.
  21. Sabés-Alsina, M.; Wallgren, M.; Sjunnesson, Y.C.; Ntallaris, T.; Lundeheim, N.; López-Béjar, M.; Morrell, J.M. Effect of season on the in vitro fertilizing ability of frozen–thawed Spanish bovine spermatozoa. J. Dairy Sci. 2020, 103, 9525–9533.
  22. Shahat, A.M.; Rizzoto, G.; Kastelic, J.P. Amelioration of heat stress-induced damage to testes and sperm quality. Theriogenology 2020, 158, 84–96.
  23. Wrzecińska, M.; Kowalczyk, A.; Kordan, W.; Cwynar, P.; Czerniawska-Piątkowska, E. Disorder of Biological Quality and Autophagy Process in Bovine Oocytes Exposed to Heat Stress and the Effectiveness of In Vitro Fertilization. Int. J. Mol. Sci. 2023, 24, 11164.
  24. Capela, L.; Leites, I.; Romao, R.; Lopes-da-Costa, L.; Pereira, R. Impact of Heat Stress on Bovine Sperm Quality and Competence. Animals 2022, 12, 975.
  25. Llamas-Luceño, N.; Hostens, M.; Mullaart, E.; Broekhuijse, M.; Lonergan, P.; Van Soom, A. High temperature-humidity index compromises sperm quality and fertility of Holstein bulls in temperate climates. J. Dairy Sci. 2020, 103, 9502–9514.
  26. Petrocchi Jasinski, F.; Evangelista, C.; Basiricò, L.; Bernabucci, U. Responses of Dairy Buffalo to Heat Stress Conditions and Mitigation Strategies: A Review. Animals 2023, 13, 1260.
  27. Papa, P.M.; Segabinazzi, L.G.; Fonseca-Alves, C.E.; Papa, F.O.; Alvarenga, M.A. Intratesticular transplantation of allogenic mesenchymal stem cells mitigates testicular destruction after induced heat stress in Miniature-horse stallions. J. Equine Vet. Sci. 2023, 132, 104961.
  28. Shakeel, M.; Yoon, M. Heat stress and stallion fertility. J. Anim. Sci. Technol. 2023, 65, 683–697.
  29. Shakeel, M.; Yoon, M. Effects of insulin-like growth factor-1 on the proliferation and apoptosis of stallion testicular cells under normal and heat stress culture conditions. Anim. Reprod. Sci. 2023, 256, 107319.
  30. Ebeid, T.A.; Aljabeili, H.S.; Al-Homidan, I.H.; Volek, Z.; Barakat, H. Ramifications of heat stress on rabbit production and role of nutraceuticals in alleviating its negative impacts: An updated review. Antioxidants 2023, 12, 1407.
  31. de Andrade, A.F.; Balogun, K.; Machaty, Z.; Knox, R.V. Effects of supplemental antioxidants on in vitro fertility measures for cryopreserved boar spermatozoa. Theriogenology 2023, 200, 33–42.
  32. Abedin, S.N.; Baruah, A.; Baruah, K.K.; Bora, A.; Dutta, D.J.; Kadirvel, G.; Katiyar, R.; Doley, S.; Das, S.; Khargharia, G.; et al. Zinc oxide and selenium nanoparticles can improve semen quality and heat shock protein expression in cryopreserved goat (Capra hircus) spermatozoa. J. Trace Elem. Med. Biol. 2023, 80, 127296.
  33. Habibi, P.; Ostad, S.N.; Heydari, A.; Monazzam, M.R.; Foroushani, A.R.; Ghazi-Khansari, M.; Golbabaei, F. Diagnostic biomarkers of heat stress induced-DNA in occupational exposure: A systematic review. J. Health Saf. Work 2023, 12, 800–819.
  34. Robinson, B.R.; Netherton, J.K.; Ogle, R.A.; Baker, M.A. Testicular heat stress, a historical perspective and two postulates for why male germ cells are heat sensitive. Biol. Rev. 2023, 98, 603–622.
  35. Abd El-Emam, M.M.; Ray, M.N.; Ozono, M.; Kogure, K. Heat stress disrupts spermatogenesis via modulation of sperm-specific calcium channels in rats. J. Therm. Biol. 2023, 112, 103465.
  36. Gan, M.; Jing, Y.; Xie, Z.; Ma, J.; Chen, L.; Zhang, S.; Zhao, Y.; Niu, L.; Wang, Y.; Li, X.; et al. Potential Function of Testicular MicroRNAs in Heat-Stress-Induced Spermatogenesis Disorders. Int. J. Mol. Sci. 2023, 24, 8809.
  37. Gao, W.J.; Li, H.X.; Feng, J.; Lu, X.R.; Yin, P.L.; Jia, H.; Ma, W.Z. Transcriptome Analysis in High Temperature Inhibiting Spermatogonial Stem Cell Differentiation In Vitro. Reprod. Sci. 2023, 30, 1938–1951.
  38. Hu, Y.; Li, Q.; Qian, Z.; Luo, K.; Luo, N. Joint Analysis of Genome-wide DNA Methylation and Transcription Sequencing Identifies the Role of BAX Gene in Heat Stress–Induced-Sertoli Cells Apoptosis. Reprod. Sci. 2024, 1–12.
  39. Liu, C.X.; Hu, S.Q.; Liu, D.L.; Xu, Y.H.; Hu, K.; Guo, J. The effect of semen cuscutae flavonoid on Sertoli cells and blood-testis barrier in male infertility: Integrating network pharmacology and experimental verification. Pharm. Biol. 2023, 61, 986–999.
  40. Wang, Y.; Wu, Z.W.; Mou, Q.; Chen, L.; Fang, T.; Zhang, Y.Q.; Yin, Z.; Du, Z.Q.; Yang, C.X. Global 3′-UTRome of porcine immature Sertoli cells altered by acute heat stress. Theriogenology 2023, 196, 79–87.
  41. Yang, J.; Ou, X.; Shu, M.; Wang, J.; Zhang, X.; Wu, Z.; Hao, W.; Zeng, H.; Shao, L. Inhibition of p38MAPK Signaling Pathway Alleviates Radiation-Induced Testicular Damage through Improving Spermatogenesis. Brit. J. Pharmacol. 2024, 181, 393–412.
  42. Zhong, L.; Zhong, H.; Dai, Y.; Zhang, Q. Effect of heat shock transcription factor 5 (Hsf5) knockdown on heat shock family in mouse Leydig cells and Sertoli cells. Basic Clin. Med. 2023, 43, 626.
  43. Deng, X.; Wang, Q.; Shi, C.; Wei, J.; Lv, Z.; Huang, S.; Duan, Y.G.; Zhang, X.; Liu, Y. Heat wave exposure and semen quality in sperm donation volunteers: A retrospective longitudinal study in south China. Environ. Res. 2023, 236, 116665.
  44. Mai, H.; Ke, J.; Li, M.; He, M.; Qu, Y.; Jiang, F.; Cai, S.; Xu, Y.; Fu, L.; Pi, L.; et al. Association of living environmental and occupational factors with semen quality in Chinese men: A cross-sectional study. Sci. Rep. 2023, 13, 15671.
  45. Tian, R.; Yang, T.; Xiao, C.; Li, F.; Fu, L.; Zhang, L.; Cai, J.; Zeng, S.; Liao, J.; Song, G.; et al. Outdoor artificial light at night and male sperm quality: A retrospective cohort study in China. Environ. Pollut. 2023, 341, 122927.
  46. Zhang, X.; Fan, Z.; Wang, Q.; Deng, X.; Xu, R.; Li, Y.; Liu, T.; Wang, R.; Shi, C.; Huang, S.; et al. Association between ambient temperature and semen quality among sperm donation volunteers in South China. Environ. Int. 2023, 173, 107809.
  47. Khan, I.; Mesalam, A.; Heo, Y.S.; Lee, S.H.; Nabi, G.; Kong, I.K. Heat Stress as a Barrier to Successful Reproduction and Potential Alleviation Strategies in Cattle. Animals 2023, 13, 2359.
  48. St-Pierre, N.R.; Cobanov, B.; Schnitkey, G. Economic losses from heat stress by US livestock industries. J. Dairy Sci. 2003, 86, E52–E77.
  49. Rhoads, M.L. Reproductive consequences of whole-body adaptations of dairy cattle to heat stress. Animal 2023, 17, 100847.
  50. Stefanska, B.; Sobolewska, P.; Fievez, V.; Pruszynska-Oszmałek, E.; Purwin, C.; Nowak, W. The impact of heat stress on performance, fertility, and adipokines involved in regulating systemic immune response during lipolysis of early lactating dairy cows. J. Dairy Sci. 2023.
  51. Barragán, A.L.; Avendaño-Reyes, L.; Mellado-Bosque, M.; Meza-Herrera, C.A.; Vicente-Pérez, R.; Castañeda, V.J.; Díaz-Molina, R.; Macías-Cruz, U. Seasonal heat stress compromises testicular thermoregulation and semen quality of Dorper rams raised in a desert climate. J. Therm. Biol. 2023, 118, 103737.
  52. Negrón-Pérez, V.M.; Fausnacht, D.W.; Rhoads, M.L. Invited review: Management strategies capable of improving the reproductive performance of heat-stressed dairy cattle. J. Dairy Sci. 2019, 102, 10695–10710.
  53. Rahman, M.B.; Schellander, K.; Luceno, N.L.; Van Soom, A. Heat stress responses in spermatozoa: Mechanisms and consequences for cattle fertility. Theriogenology 2018, 113, 102–112.
  54. de Aguiar, L.H.; Hyde, K.A.; Pedroza, G.H.; Denicol, A.C. Heat stress impairs in vitro development of preantral follicles of cattle. Anim. Reprod. Sci. 2020, 213, 106277.
  55. Khan, A.; Dou, J.; Wang, Y.; Jiang, X.; Khan, M.Z.; Luo, H.; Usman, T.; Zhu, H. Evaluation of heat stress effects on cellular and transcriptional adaptation of bovine granulosa cells. J. Anim. Sci. Biotechnol. 2020, 11, 25.
  56. Alemu, T.W.; Pandey, H.O.; Wondim, D.S.; Gebremedhn, S.; Neuhof, C.; Tholen, E.; Holker, M.; Schellander, K.; Tesfaye, D. Oxidative and endoplasmic reticulum stress defense mechanisms of bovine granulosa cells exposed to heat stress. Theriogenology 2018, 110, 130–141.
  57. De Rensis, F.; Lopez-Gatius, F.; García-Ispierto, I.; Morini, G.; Scaramuzzi, R.J. Causes of declining fertility in dairy cows during the warm season. Theriogenology 2017, 91, 145–153.
  58. Schüller, L.K.; Michaelis, I.; Heuwieser, W. Impact of heat stress on estrus expression and follicle size in estrus under field conditions in dairy cows. Theriogenology 2017, 102, 48–53.
  59. Ho, K.T.; Balboula, A.Z.; Homma, K.; Takanari, J.; Bai, H.; Kawahara, M.; Nguyen, K.T.; Takahashi, M. Synergistic effect of standardized extract of Asparagus officinalis stem and heat shock on progesterone synthesis with lipid droplets and mitochondrial function in bovine granulosa cells. J. Steroid Biochem. Mol. Biol. 2023, 225, 106181.
  60. Menjivar, N.G.; Gad, A.; Gebremedhn, S.; Ghosh, S.; Tesfaye, D. Granulosa cell-derived extracellular vesicles mitigate the detrimental impact of thermal stress on bovine oocytes and embryos. Front. Cell Dev. Biol. 2023, 11, 1142629.
  61. Miękiewska, K.; Kordowitzki, P.; Pareek, C.S. Effects of Heat Stress on Bovine Oocytes and Early Embryonic Development—An Update. Cells 2022, 11, 4073.
  62. Yüzen, D.; Graf, I.; Diemert, A.; Arck, P.C. Climate change and pregnancy complications: From hormones to the immune response. Front. Endocrinol. 2023, 14, 1149284.
  63. Chotimanukul, S.; Suwimonteerabutr, J.; Techakumphu, M.; Swangchan-Uthai, T. In vitro effects of short-term and long-term heat exposures on the immune response and prostaglandin biosynthesis in bovine endometrial cells. Animals 2022, 12, 2359.
  64. Roth, Z. Heat stress reduces maturation and developmental capacity in bovine oocytes. Reprod. Fertil. Dev. 2021, 33, 66–75.
  65. Roth, Z. Influence of heat stress on reproduction in dairy cows—Physiological and practical aspects. J. Anim. Sci. 2020, 98, S80–S87.
  66. Roth, Z. Reproductive physiology and endocrinology responses of cows exposed to environmental heat stress-Experiences from the past and lessons for the present. Theriogenology 2020, 155, 150–156.
  67. Huber, E.; Notaro, U.S.; Recce, S.; Rodríguez, F.M.; Ortega, H.H.; Salvetti, N.R.; Rey, F. Fetal programming in dairy cows: Effect of heat stress on progeny fertility and associations with the hypothalamic-pituitary-adrenal axis functions. Anim. Reprod. Sci. 2020, 216, 106348.
  68. Nanas, I.; Chouzouris, T.M.; Dadouli, K.; Dovolou, E.; Stamperna, K.; Barbagianni, M.; Valasi, I.; Tsiaras, A.; Amiridis, G.S. A study on stress response and fertility parameters in phenotypically thermotolerant and thermosensitive dairy cows during summer heat stress. Reprod. Domest. Anim. 2020, 55, 1774–1783.
  69. Payton, R.R.; Rispoli, L.A.; Nagle, K.A.; Gondro, C.; Saxton, A.M.; Voy, B.H.; Edwards, J.L. Mitochondrial-related consequences of heat stress exposure during bovine oocyte maturation persist in early embryo development. J. Reprod. Dev. 2018, 64, 243–251.
  70. Diaz, F.A.; Gutierrez-Castillo, E.J.; Foster, B.A.; Hardin, P.T.; Bondioli, K.R.; Jiang, Z. Evaluation of seasonal heat stress on transcriptomic profiles and global DNA methylation of bovine oocytes. Front. Genet. 2021, 12, 699920.
  71. Gendelman, M.; Roth, Z. In vivo vs. in vitro models for studying the effects of elevated temperature on the GV-stage oocyte, subsequent developmental competence and gene expression. Anim. Reprod. Sci. 2012, 134, 125–134.
  72. Gendelman, M.; Roth, Z. Incorporation of coenzyme Q10 into bovine oocytes improves mitochondrial features and alleviates the effects of summer thermal stress on developmental competence. Biol. Reprod. 2012, 87, 118.
  73. Makris, A.; Alevra, A.I.; Exadactylos, A.; Papadopoulos, S. The Role of Melatonin to Ameliorate Oxidative Stress in Sperm Cells. Int. J. Mol. Sci. 2023, 24, 15056.
  74. Zhong, R.Z.; Zhou, D.W. Oxidative Stress and Role of Natural Plant-Derived Antioxidants in Animal Reproduction. J. Integr. Agric. 2013, 12, 1826–1838.
  75. Mohlala, K.; Offor, U.; Monageng, E.; Takalani, N.B.; Opuwari, C.S. Overview of the Effects of Moringa oleifera Leaf Extract on Oxidative Stress and Male Infertility: A Review. Appl. Sci. 2023, 13, 4387.
  76. Shahat, A.M.; Thundathil, J.C.; Kastelic, J.P. Melatonin improves post-thaw sperm quality after mild testicular heat stress in rams. Reprod. Domest. Anim. 2023, 58, 423–430.
  77. El-Gindy, Y.M.; Sabir, S.A.; Zahran, S.M.; Morshedy, S.A. The protective effect of aqueous orange peel extract against severe heat stress on reproductive efficiency, milk yield, and antioxidant status of female rabbits. J. Therm. Biol. 2023, 111, 103403.
  78. El-Gindy, Y.M.; Sabir, S.A.; Zahran, S.M.; Ahmed, M.H.; Reuben, R.C.; Salem, A.Z. Effect of dietary onion (Allium cepa L.) powder as an antioxidant on semen quality, blood biochemicals, and reproductive parameters, as well as immunological variables of rabbit bucks under severe heat stress. Trop. Anim. Health Prod. 2023, 55, 380.
  79. El-Gindy, Y.M.; Zahran, S.M.; Ahmed, M.H.; Adegbeye, M.J.; Salem, A.Z.; Salam, M.Y. Enhancing semen quality, antioxidant status and sex hormones of V-line rabbit bucks fed on supplemented diets with dried moringa leaves. Anim. Biotechnol. 2023, 34, 2626–2635.
  80. El-Ratel, I.T.; El-Kholy, K.H.; Mousa, N.A.; El-Said, E.A. Impacts of selenium nanoparticles and spirulina alga to alleviate the deleterious effects of heat stress on reproductive efficiency, oxidative capacity and immunity of doe rabbits. Anim. Biotechnol. 2023, 34, 3519–3532.
  81. Jimoh, O.A.; Daramola, O.T.; Okin-Aminu, H.O.; Ojo, O.A.; Oyeyemi, W.A. Effect of phytogenic supplements on the reproductive physiology and metabolic hormones of rabbits exposed to heat stress conditions. J. Therm. Biol. 2023, 112, 103438.
  82. Naspinska, R.; Moreira da Silva, M.H.; Moreira da Silva, F. Current Advances in Bovine In Vitro Maturation and Embryo Production Using Different Antioxidants: A Review. J. Dev. Biol. 2023, 11, 36.
  83. Toosinia, S.; Davoodian, N.; Arabi, M.; Kadivar, A. Ameliorating Effect of Sodium Selenite on Developmental and Molecular Response of Bovine Cumulus-Oocyte Complexes Matured In Vitro under Heat Stress Condition. Biol. Trace Elem. Res. 2024, 202, 161–174.
  84. Tripathi, S.K.; Nandi, S.; Gupta, P.S.; Mondal, S. Antioxidants supplementation improves the quality of in vitro produced ovine embryos with amendments in key development gene expressions. Theriogenology 2023, 201, 41–52.
  85. Yaacobi-Artzi, S.; Shimoni, C.; Kalo, D.; Hansen, P.J.; Roth, Z. Melatonin slightly alleviates the effect of heat shock on bovine oocytes and resulting blastocysts. Theriogenology 2020, 158, 477–489.
  86. Rakha, S.I.; Elmetwally, M.A.; El-Sheikh Ali, H.; Balboula, A.; Mahmoud, A.M.; Zaabel, S.M. Importance of Antioxidant Supplementation during In Vitro Maturation of Mammalian Oocytes. Vet. Sci. 2022, 9, 439.
  87. Zhang, B.; Pan, C.; Feng, C.; Yan, C.; Yu, Y.; Chen, Z.; Guo, C.; Wang, X. Role of mitochondrial reactive oxygen species in homeostasis regulation. Redox Rep. 2022, 27, 45–52.
  88. Aitken, R.J.; Drevet, J.R.; Moazamian, A.; Gharagozloo, P. Male infertility and oxidative stress: A focus on the underlying mechanisms. Antioxidants 2022, 11, 306.
  89. Juan, C.A.; Pérez de la Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The chemistry of reactive oxygen species (ROS) revisited: Outlining their role in biological macromolecules (DNA, lipids and proteins) and induced pathologies. Int. J. Mol. Sci. 2021, 22, 4642.
  90. Belhadj Slimen, I.; Najar, T.; Ghram, A.; Dabbebi, H.; Ben Mrad, M.; Abdrabbah, M. Reactive oxygen species, heat stress and oxidative-induced mitochondrial damage. A review. Int. J. Hyperth. 2014, 30, 513–523.
  91. Bisht, S.; Faiq, M.; Tolahunase, M.; Dada, R. Oxidative stress and male infertility. Nat. Rev. Urol. 2017, 14, 470–485.
  92. Drevet, J.R.; Hallak, J.; Nasr-Esfahani, M.H.; Aitken, R.J. Reactive oxygen species and their consequences on the structure and function of mammalian spermatozoa. Antioxid. Redox Signal. 2022, 37, 481–500.
  93. Sharma, P.; Kaushal, N.; Saleth, L.R.; Ghavami, S.; Dhingra, S.; Kaur, P. Oxidative stress-induced apoptosis and autophagy: Balancing the contrary forces in spermatogenesis. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2023, 1869, 166742.
  94. Aldahhan, R.A.; Stanton, P.G.; Ludlow, H.; de Kretser, D.M.; Hedger, M.P. Acute heat-treatment disrupts inhibin-related protein production and gene expression in the adult rat testis. Mol. Cell. Endocrinol. 2019, 498, 110546.
  95. Khan, A.; Khan, M.Z.; Dou, J.; Xu, H.; Liu, L.; Zhu, H.; Wang, Y. SOD1 Gene silencing promotes apoptosis and suppresses proliferation of heat-stressed bovine granulosa cells via induction of oxidative stress. Vet. Sci. 2021, 8, 326.
  96. Agarwal, A.; Mulgund, A.; Hamada, A.; Chyatte, M.R. A unique view on male infertility around the globe. Reprod. Biol. Endocrinol. 2015, 13, 37.
  97. Henriques, M.C.; Santiago, J.; Patrício, A.; Herdeiro, M.T.; Loureiro, S.; Fardilha, M. Smoking Induces a Decline in Semen Quality and the Activation of Stress Response Pathways in Sperm. Antioxidants 2023, 12, 1828.
  98. Li, H.; Cong, X.; Yu, W.; Jiang, Z.; Fu, K.; Cao, R.; Tian, W.; Feng, Y. Baicalin inhibits oxidative injures of mouse uterine tissue induced by acute heat stress through activating the Keap1/Nrf2 signaling pathway. Res. Vet. Sci. 2022, 152, 717–725.
  99. Cai, D.; Li, X.; Xu, Q.; Li, H.; Liu, R.; Chen, J.; Jiang, X.; Sun, J.; Lai, C.; Bai, W. Cyanidin-3-O-glucoside and protocatechuic acid alleviate heat stress-induced testicular damage. Food Funct. 2023, 14, 2200–2211.
  100. Mbegbu, E.C.; Ikele, C.G.; Obidike, I.R.; Parrish, J.J. Immunomodulatory potentials of saccharomyces cerevisiae in mitigation of testicular heat stress alterations. J. Reprod. Immunol. 2023, 159, 104057.
  101. Wang, K.; Li, Z.; Li, Y.; Li, X.; Suo, Y.; Li, C. Impacts of elevated temperature on morphology, oxidative stress levels, and testosterone synthesis in ex vivo cultured porcine testicular tissue. Theriogenology 2023, 212, 181–188.
  102. Cai, H.; Qin, D.; Peng, S. Responses and coping methods of different testicular cell types to heat stress: Overview and perspectives. Biosci. Rep. 2021, 41, BSR20210443.
  103. Wang, C.; He, C.; Gao, Y.; Wang, K.; Liang, M. Heat exposure promotes apoptosis and pyroptosis in Sertoli cells. Biocell 2023, 47, 155–164.
  104. Deng, C.C.; Zhang, J.P.; Huo, Y.N.; Xue, H.Y.; Wang, W.; Zhang, J.J.; Wang, X.Z. Melatonin alleviates the heat stress-induced impairment of Sertoli cells by reprogramming glucose metabolism. J. Pineal Res. 2022, 73, e12819.
  105. Xue, H.; Huo, Y.; Hu, Y.; Zhang, J.; Deng, C.; Zhang, J.; Wang, X. The role of ALOX15B in heat stress-induced apoptosis of porcine Sertoli cells. Theriogenology 2022, 185, 6–15.
  106. Hu, Y.; Hu, H.; Yin, L.; Wang, L.; Luo, K.; Luo, N. Arachidonic acid impairs the function of the blood-testis barrier via triggering mitochondrial complex-ROS-P38 MAPK axis in hyperthermal Sertoli cells. Ecotoxicol. Environ. Saf. 2023, 252, 114598.
  107. He, C.; Sun, J.; Yang, D.; He, W.; Wang, J.; Qin, D.; Zhang, H.; Cai, H.; Liu, Y.; Li, N.; et al. Nrf2 activation mediates the protection of mouse Sertoli Cells damage under acute heat stress conditions. Theriogenology 2022, 177, 183–194.
  108. Monageng, E.; Offor, U.; Takalani, N.B.; Mohlala, K.; Opuwari, C.S. A Review on the Impact of Oxidative Stress and Medicinal Plants on Leydig Cells. Antioxidants 2023, 12, 1559.
  109. Xiong, Y.; Li, J.; He, S. Zinc Protects against Heat Stress–Induced Apoptosis via the Inhibition of Endoplasmic Reticulum Stress in TM3 Leydig Cells. Biol. Trace Elem. Res. 2022, 200, 728–739.
  110. Kawano, K.; Sakaguchi, K.; Madalitso, C.; Ninpetch, N.; Kobayashi, S.; Furukawa, E.; Yanagawa, Y.; Katagiri, S. Effect of heat exposure on the growth and developmental competence of bovine oocytes derived from early antral follicles. Sci. Rep. 2022, 12, 8857.
  111. Wang, Y.R.; Chen, K.L.; Li, C.M.; Li, L.; Wang, G.L. Heme oxygenase 1 regulates apoptosis induced by heat stress in bovine ovarian granulosa cells via the ERK1/2 pathway. J. Cell. Physiol. 2019, 234, 3961–3972.
  112. De Rensis, F.; Saleri, R.; Garcia-Ispierto, I.; Scaramuzzi, R.; López-Gatius, F. Effects of heat stress on follicular physiology in dairy cows. Animals 2021, 11, 3406.
  113. Li, H.; Cong, X.; Sui, J.; Jiang, Z.; Fu, K.; Huan, Y.; Cao, R.; Tian, W.; Feng, Y. Baicalin enhances the thermotolerance of mouse blastocysts by activating the ERK1/2 signaling pathway and preventing mitochondrial dysfunction. Theriogenology 2022, 178, 85–94.
  114. Sammad, A.; Luo, H.; Hu, L.; Zhu, H.; Wang, Y. Transcriptome reveals granulosa cells coping through redox, inflammatory and metabolic mechanisms under acute heat stress. Cells 2022, 11, 1443.
  115. Cardone, D.A.; Cáceres, A.R.; Sanhueza, M.A.; Bruna, F.A.; Laconi, M.R. Effects of short-term in vitro heat stress on bovine preantral follicles. Livest. Sci. 2022, 264, 105076.
  116. Zhang, S.X.; Wang, D.L.; Qi, J.J.; Yang, Y.W.; Sun, H.; Sun, B.X.; Liang, S. Chlorogenic acid ameliorates the heat stress-induced impairment of porcine Sertoli cells by suppressing oxidative stress and apoptosis. Theriogenology 2024, 214, 148–156.
  117. Bai, L.; Zhang, Y.; Zheng, C.; Xu, S.; He, Y.; Yu, G.; Huang, D.; Huang, Y.; Li, M.; Xu, C. Tanshinone IIA protects mouse testes from heat stress injury by inhibiting apoptosis and TGFβ1/Smad2/Smad3 signaling pathway. Cell Stress Chaperones 2023, 28, 749–759.
  118. Xiong, Y.; Jin, E.; Yin, Q.; Che, C.; He, S. Boron attenuates heat stress–induced apoptosis by inhibiting endoplasmic reticulum stress in mouse granulosa cells. Biol. Trace Elem. Res. 2021, 199, 611–621.
  119. Stamperna, K.; Giannoulis, T.; Dovolou, E.; Kalemkeridou, M.; Nanas, I.; Dadouli, K.; Moutou, K.; Mamuris, Z.; Amiridis, G.S. Heat Shock Protein 70 improves in vitro embryo yield and quality from heat stressed bovine oocytes. Animals 2021, 11, 1794.
  120. Jais, A.; Einwallner, E.; Sharif, O.; Gossens, K.; Lu, T.T.; Soyal, S.M.; Medgyesi, D.; Neureiter, D.; Paier-Pourani, J.; Dalgaard, K.; et al. Heme oxygenase-1 drives metaflammation and insulin resistance in mouse and man. Cell 2014, 158, 25–40.
  121. Bindu, S.; Pal, C.; Dey, S.; Goyal, M.; Alam, A.; Iqbal, M.S.; Dutta, S.; Sarkar, S.; Kumar, R.; Maity, P.; et al. Translocation of heme oxygenase-1 to mitochondria is a novel cytoprotective mechanism against non-steroidal anti-inflammatory drug-induced mitochondrial oxidative stress, apoptosis, and gastric mucosal injury. J. Biol. Chem. 2011, 286, 39387–39402.
  122. Kondo, R.; Gleixner, K.V.; Mayerhofer, M.; Vales, A.; Gruze, A.; Samorapoompichit, P.; Greish, K.; Krauth, M.T.; Aichberger, K.J.; Pickl, W.F.; et al. Identification of heat shock protein 32 (Hsp32) as a novel survival factor and therapeutic target in neoplastic mast cells. Blood 2007, 110, 661–669.
  123. Zenclussen, M.L.; Jensen, F.; Rebelo, S.; El-Mousleh, T.; Casalis, P.A.; Zenclussen, A.C. Heme oxygenase-1 expression in the ovary dictates a proper oocyte ovulation, fertilization, and corpora lutea maintenance. Am. J. Reprod. Immunol. 2012, 67, 376–382.
  124. Wang, Y.; Yang, C.; Nahla Abdalla Hassan, E.; Li, C.; Yang, F.; Wang, G.; Li, L. HO-1 reduces heat stress-induced apoptosis in bovine granulosa cells by suppressing oxidative stress. Aging 2019, 11, 5535.
  125. Faheem, M.S.; Dessouki, S.M.; Abdel-Rahman, F.E.; Ghanem, N. Physiological and molecular aspects of heat-treated cultured granulosa cells of Egyptian buffalo (Bubalus bubalis). Anim. Reprod. Sci. 2021, 224, 106665.
  126. Faheem, M.S.; Ghanem, N.; Gad, A.; Procházka, R.; Dessouki, S.M. Adaptive and Biological Responses of Buffalo Granulosa Cells Exposed to Heat Stress under In Vitro Condition. Animals 2021, 11, 794.
  127. Guan, J.Y.; Liao, T.T.; Yu, C.L.; Luo, H.Y.; Yang, W.R.; Wang, X.Z. ERK1/2 regulates heat stress-induced lactate production via enhancing the expression of HSP70 in immature boar Sertoli cells. Cell Stress Chaperones 2018, 23, 1193–1204.
  128. Liang, Y.; Liu, J.; Feng, Z. The regulation of cellular metabolism by tumor suppressor p53. Cell Biosci. 2013, 3, 9.
  129. Cui, W.; Li, B.; Bai, Y.; Miao, X.; Chen, Q.; Sun, W.; Tan, Y.; Luo, P.; Zhang, C.; Zheng, S.; et al. Potential role for Nrf2 activation in the therapeutic effect of MG132 on diabetic nephropathy in OVE26 diabetic mice. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E87–E99.
  130. Cullinan, S.B.; Gordan, J.D.; Jin, J.; Harper, J.W.; Diehl, J.A. The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: Oxidative stress sensing by a Cul3-Keap1 ligase. Mol. Cell. Biol. 2004, 24, 8477–8486.
  131. Pankiv, S.; Clausen, T.H.; Lamark, T.; Brech, A.; Bruun, J.-A.; Outzen, H.; Øvervatn, A.; Bjørkøy, G.; Johansen, T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 2007, 282, 24131–24145.
  132. Lau, A.; Zheng, Y.; Tao, S.; Wang, H.; Whitman, S.A.; White, E.; Zhang, D.D. Arsenic inhibits autophagic flux, activating the Nrf2-Keap1 pathway in a p62-dependent manner. Mol. Cell. Biol. 2013, 33, 2436–2446.
  133. Lau, A.; Wang, X.-J.; Zhao, F.; Villeneuve, N.F.; Wu, T.; Jiang, T.; Sun, Z.; White, E.; Zhang, D.D. A noncanonical mechanism of Nrf2 activation by autophagy deficiency: Direct interaction between Keap1 and p62. Mol. Cell. Biol. 2010, 30, 3275–3285.
  134. Copple, I.M.; Lister, A.; Obeng, A.D.; Kitteringham, N.R.; Jenkins, R.E.; Layfield, R.; Foster, B.J.; Goldring, C.E.; Park, B.K. Physical and functional interaction of sequestosome 1 with Keap1 regulates the Keap1-Nrf2 cell defense pathway. J. Biol. Chem. 2010, 285, 16782–16788.
  135. Li, Z.; Li, Y.; Zhou, X.; Dai, P.; Li, C. Autophagy involved in the activation of the Nrf2-antioxidant system in testes of heat-exposed mice. J. Therm. Biol. 2018, 71, 142–152.
  136. Rotimi, D.E.; Ojo, O.A.; Olaolu, T.D.; Adeyemi, O.S. Exploring Nrf2 as a therapeutic target in testicular dysfunction. Cell Tissue Res. 2022, 390, 23–33.
  137. Li, Y.; Cao, Y.; Wang, F.; Pu, S.; Zhang, Y.; Li, C. Tert-butylhydroquinone attenuates scrotal heat-induced damage by regulating Nrf2-antioxidant system in the mouse testis. Gen. Comp. Endocrinol. 2014, 208, 12–20.
  138. Li, Y.; Cao, Y.; Wang, F.; Li, C. Scrotal heat induced the Nrf2-driven antioxidant response during oxidative stress and apoptosis in the mouse testis. Acta Histochem. 2014, 116, 883–890.
  139. Li, Y.; Huang, Y.; Piao, Y.; Nagaoka, K.; Watanabe, G.; Taya, K.; Li, C.M. Protective effects of nuclear factor erythroid 2-related factor 2 on whole body heat stress-induced oxidative damage in the mouse testis. Reprod. Biol. Endocrinol. 2013, 11, 23.
  140. Sui, J.; Feng, Y.; Li, H.; Cao, R.; Tian, W.; Jiang, Z. Baicalin protects mouse testis from injury induced by heat stress. J. Therm. Biol. 2019, 82, 63–69.
  141. Feng, J.; He, Y.; Shen, Y.; Zhang, G.; Ma, S.; Zhao, X.; Zhang, Y. Protective effects of nuclear factor erythroid 2-related factor on oxidative stress and apoptosis in the testis of mice before adulthood. Theriogenology 2020, 148, 112–121.
  142. Murata, H.; Kunii, H.; Kusama, K.; Sakurai, T.; Bai, H.; Kawahara, M.; Takahashi, M. Heat stress induces oxidative stress and activates the KEAP1-NFE2L2-ARE pathway in bovine endometrial epithelial cells. Biol. Reprod. 2021, 105, 1114–1125.
  143. Li, J.; Zhao, W.; Zhu, J.; Ju, H.; Liang, M.; Wang, S.; Chen, S.; Ferreira-Dias, G.; Liu, Z. Antioxidants and Oxidants in Boar Spermatozoa and Their Surrounding Environment Are Associated with AMPK Activation during Liquid Storage. Vet. Sci. 2023, 10, 214.
  144. Froment, P.; Plotton, I.; Giulivi, C.; Fabre, S.; Khoueiry, R.; Mourad, N.I.; Horman, S.; Rame, C.; Rouillon, C.; Grandhaye, J.; et al. At the crossroads of fertility and metabolism: The importance of AMPK-dependent signaling in female infertility associated with hyperandrogenism. Hum. Reprod. 2022, 37, 1207–1228.
  145. Yang, W.; Wang, L.; Wang, F.; Yuan, S. Roles of AMP-activated protein kinase (AMPK) in mammalian reproduction. Front. Cell Dev. Biol. 2020, 8, 593005.
  146. Meroni, S.B.; Galardo, M.N.; Rindone, G.; Gorga, A.; Riera, M.F.; Cigorraga, S.B. Molecular mechanisms and signaling pathways involved in sertoli cell proliferation. Front. Endocrinol. 2019, 10, 224.
  147. Ni, F.D.; Hao, S.L.; Yang, W.X. Multiple signaling pathways in Sertoli cells: Recent findings in spermatogenesis. Cell Death Dis. 2019, 10, 541.
  148. Yang, W.R.; Liao, T.T.; Bao, Z.Q.; Zhou, C.Q.; Luo, H.Y.; Lu, C.; Pan, M.H.; Wang, X.Z. Role of AMPK in the expression of tight junction proteins in heat-treated porcine Sertoli cells. Theriogenology 2018, 121, 42–52.
  149. Bertoldo, M.J.; Guibert, E.; Faure, M.; Guillou, F.; Ramé, C.; Nadal-Desbarats, L.; Foretz, M.; Viollet, B.; Dupont, J.; Froment, P. Specific deletion of AMP-activated protein kinase (α1AMPK) in mouse Sertoli cells modifies germ cell quality. Mol. Cell. Endocrinol. 2016, 423, 96–112.
  150. Xiao, L.; Fang, Z.; Wang, Q.; Sheng, X.; Qi, X.; Xing, K.; Guo, Y.; Ni, H.; Wang, X.; Zhang, Y. Curcumin Ameliorates Age-Induced Tight Junction Impaired in Porcine Sertoli Cells by Inactivating the NLRP3 Inflammasome through the AMPK/SIRT3/SOD2/mtROS Signaling Pathway. Oxid. Med. Cell. Longev. 2023, 2023, 1708251.
More
Video Production Service