Indian Journal of Animal Research

  • Chief EditorK.M.L. Pathak

  • Print ISSN 0367-6722

  • Online ISSN 0976-0555

  • NAAS Rating 6.40

  • SJR 0.263

  • Impact Factor 0.4 (2024)

Frequency :
Monthly (January, February, March, April, May, June, July, August, September, October, November and December)
Indexing Services :
Science Citation Index Expanded, BIOSIS Preview, ISI Citation Index, Biological Abstracts, Scopus, AGRICOLA, Google Scholar, CrossRef, CAB Abstracting Journals, Chemical Abstracts, Indian Science Abstracts, EBSCO Indexing Services, Index Copernicus

Role of Caspase 3, Caspase 8 and Caspase 9 in Male Infertility and Sperm Cryoinjury: A Review

Anna M. Shitikova1,2,*, Mikhail M. Atroshchenko2, Marya G. Engalycheva1, Eduard S. Belskikh1
  • 0000-0002-4004-9058, 0000-0001-6023-0332, 0000-0002-9719-036X, 0000-0003-1803-0542
1Ryazan State Medical University, 9 Vysokovoltnaya str., Ryazan, 390026, Russia.
2All-Russian Research Institute for Horse Breeding (ARRIH), Ryazan Region, Rybnovskij District, Divovo, 391105, Russia.

Apoptosis is an integral part of sperm development, quality control and differentiation. Caspases are proteolytic enzymes that play a key role in apoptosis. The aim of this review article is to analyze the role of caspases 3, 8 and 9 in male infertility and sperm cryopreservation. The review also discusses the effect of cryopreservation on the activation of caspases in sperm, leading to apoptosis and decreased sperm quality. Finally, the article explores the use of caspase inhibitors as a strategy to improve sperm cryopreservation protocols and preserve sperm quality. Caspase 3 is the dominant effector caspase, while caspase 8 and 9 are involved in the extrinsic and intrinsic apoptotic pathways, respectively. Increased activity of these caspases has been associated with various sperm quality issues, such as teratozoospermia, asthenozoospermia, oligozoospermia and reduced cryostability. The review emphasizes the importance of understanding the role of caspases in sperm apoptosis and their potential as diagnostic markers and therapeutic targets for male infertility and sperm cryopreservation.

Despite the fact that the biological processes of spermato-genesis and sperm function have been well studied in mammals, research in the field of male reproductive health does not lose its relevance every year. These studies have not only a fundamental basis that complements the set of knowledge about the sperm biology, but also an important practical and social significance. The search for new biochemical markers of male fertility disorders can make a significant contribution to understanding the molecular foundations of male infertility. Study of sperm apoptosis can help in regulating the number and quality of germ cells (Asadi et al., 2021; Said et al., 2004; Zalata et al., 2016). In addition, it is important to consider the role of these studies to improve cryopreservation protocols, since this technology has been widely used not only to increase human reproductive capabilities (Said et al., 2010), but also is of great importance for preserving the genetic material of valuable animal breeds (Atroshchenko et al., 2019; Contreras et al., 2023; Upadhyay et al., 2021). Undoubtedly, the reproductive potential of the animals need to be exploited to its maximum to achieve optimum production in a herd (Ahanger et al., 2024; Chandra et al., 2011; Madkar et al., 2022). We also see particular interest in the search for biochemical markers of seminal plasma that can predict the outcome of cryopreservation (Atroschenko et al., 2022, Shitikova et al., 2023, Shitikova et al., 2024), while partici-pants in apoptotic events are ideal candidates for this role.
 
Apoptosis in spermatozoa
 
Apoptosis is an integral part of sperm development, quality control and differentiation (Asadi, 2021). In the postnatal period, it is responsible for the death and phagocytosis of premeiotic germ cells in order to bring their number in line with the number of Sertoli cells, as well as for the removal of damaged and abnormal germ cells during active spermatogenesis in the post-puberty period. Genetic and exogenous damages affecting spermatogenesis enhance the endogenous mechanism of germ cell apoptosis and can lead to insufficient or absent sperm production (Murphy et al., 2014). It has been shown that germ cell apoptosis increases after testicular damage, such as exposure to toxic substances, varicocele, testicular torsion, hormonal deprivation and genetic abnormalities, as well as the freeze-thaw reaction (Almeida et al., 2005). Cryopreservation of spermatozoa causes lipid peroxidation, free radical production, formation of intracellular and extracellular ice crystals, dehydration, membrane damage, mitochondrial dysfunction, DNA fragmentation, cytoskeletal disorders which, in turn, can trigger a cascade of apoptotic events in humans (Said, 2010), bulls (Upadhyay et al., 2021), stallions (Atroshchenko et al., 2019; Contreras et al., 2023). Cryopreservation causes death of spermatozoa using both the external and internal pathways of apoptosis (Savitskaya and Onishchenko, 2016).
 
Caspases
 
An important role in the process of apoptosis is assigned to cysteine proteases - caspases, calpains and cathepsins. Cathepsins act in the acidic environment of lysosomes and the acrosome of spermatozoa (Moreno and Alvarado, 2006) and perform many different biological functions, which has fueled the interest of researchers in it for many years (Fomina and Kudlaeva, 2016; Fomina et al., 2017). While cathepsins may serve as potential indicators of cryostability in seminal plasma (Shitikova et al., 2023), our current review will concentrate specifically on caspases.

Caspases (cysteine-dependent aspartate specific protease) are proteolytic enzymes that cleave proteins at the locations of the amino acid aspartate (Shalini et al., 2015). These are divided into initiatory, inflammatory and effector (executive) ones. All caspases are initially synthesized as inactive precursors (procaspases) and are activated as needed by initiator caspases due to cleavage of a small portion of the molecule. Initiatory caspases, in turn, are activated in a more complex way - by special protein complexes (apoptosomes, PIDD-osomes, DISC) (Man et al., 2017; Van Opdenbosch and Lamkanfi 2019).

To date, it has been established that caspases 2, 8, 9 and 10 are initiatory, 3, 6 and 7 are effector, 1, 4, 5, 11, 12, 13 are inflammatory (Kesavardhana et al., 2020). When an apoptotic stimulus appears, caspase initiators get activated, triggering the biochemical cascade of apoptosis. Caspase initiator 8 plays the most important role in the extrinsic pathway of apoptosis and activated caspase-9 plays a key role in the intrinsic pathway of apoptosis, cleaves underlying caspases such as caspase-3, -6 and -7, initiating a cascade of caspases. Activation of caspase-3 marks the point of no return in the process of apoptosis and is responsible for the cleavage of key proteins, leading to the final destruction of cells (Chowdhury et al., 2008). Considering the above facts study of caspases 3, 8 and 9 is extremely essential.
 
Caspase 3
 
Caspase 3 (CASP3, CPP32/Yama/apopain) is a proteolytic enzyme consisting of 277 amino acid residues, with a molecular weight of 31.608 kDa. Like all caspases, CASP3 is synthesized as an inactive proenzyme, which is proteoly-tically processed in conservative asparagine residues to form two subunits, large and small, which dimerize to form an active enzyme. The dimerized enzyme cleaves and activates caspases 6 and 7; and the enzyme itself is modified and activated by caspases 8, 9 and 10 (Wyllie, 1997; Crowley and Waterhouse, 2016).

The active center of the enzyme contains a cysteine residue (Cys-163) and a histidine residue (His-121), which stabilize the cleavage of the peptide bond in the protein molecule at the C-terminus at the location of aspartic acid when it is part of a specific 4-amino acid sequence, recognizing the tetrapeptide motif Asp-x-x-Asp (Agniswamy et al., 2007).  

Caspase 3 is active in a wide pH range, which is slightly higher (more basic) than many other caspase effectors. This wide range indicates that caspase 3 can be fully active under normal and apoptotic conditions (Stennicke and Salvesen, 1997).

Caspase 3 is activated in the apoptotic cell by both extrinsic and intrinsic pathways (Ghavami et al., 2009). A distinctive feature of the caspase 3 zymogen is the need for strict regulation, because if it is not regulated, excessive caspase activity leads to total cell death (both healthy and pathological cells) (Boatright and Salvesen, 2003). As a caspase effector, the caspase-3 zymogen has practically no activity until it is cleaved by initiator caspase after transmission of a special apoptotic signal (Walters et al., 2009). External activation triggers a caspase cascade characteristic of the apoptotic pathway, in which caspase 3 plays a dominant role. During the internal activation of caspase, cytochrome C molecules from mitochondria work in combination with caspase 9, apoptosis-activating factor 1 (APAF1) and ATP necessary for the enzyme. The development of oxidative distress and mitochondria damage is considered one of the main reasons for the release of cytochrome C from mitochondria (Li et al., 2004; Shalini et al., 2015). 

One of the ways to inhibit CASP3 is to use the IAP family of proteins (apoptosis inhibitors), which includes c-IAP1, c-IAP2, XIAP and ML-IAP (Lavrik et al., 2005; Poreba et al., 2013).
 
Caspase 3 in sperm
 
It has been established that caspases-3, 8, 9 are present in both sperm and seminal plasma. The level of caspase-3 and caspase-8 in human seminal plasma is significantly higher than in sperm, so it makes sense to investigate the activity of caspases both in the spermatozoa themselves and in the seminal plasma. In vitro sperm processing methods (centrifugal force during mechanical processing, magnetic field strength during MAC separation, cooling during deep hypothermia, centrifugation in a density gradient) have a certain effect on spermatozoa, therefore it is difficult to determine whether caspase is activated before or after ejaculation or simultaneously. Provided that caspase is present in the seminal plasma, the effect due to in vitro treatment can be ignored and the state of sperm apoptosis before ejaculation can be reflected directly. Caspases isolated from spermatozoa may reflect apoptosis after ejaculation (Wei et al., 2015). 

Almeida et al. (2005) found that the activity of caspase-3 of sperm cells in humans is apparently associated with teratozoospermia and asthenozoospermia. This is due to the fact that nuclear, mitochondrial and cytoskeletal abnormalities cause activation of caspase-3 during spermiogenesis or maturation of spermatozoa (Almeida et al., 2005). Xu et al., (2020) found that miR-210-3p (microRNA) induces apoptosis of spermatogenic cells, contributing to the activation of caspase-3. The activity of caspase-3 was detected in sperm midpiece region and it was shown that it is largely associated with low sperm motility (Weng et al., 2002) or with reduced normal sperm concentration, motility and morphology (Wang et al., 2003). Ejaculate with a large number of caspases positive spermatozoa negatively correlates with the rate of fertilization after in vitro fertilization (Marchetti et al., 2004). It has been experimentally established that thermal exposure to the scrotum, especially for a long time, increases the activity of caspase 3 (Zhang et al., 2015). Increased activity of caspase-3 was found in Sertoli cells of patients with Sertoli cell-only syndrome suggesting that apoptosis might be an active mechanism in this syndrome. Hypospermatogenesis and oligozoospermia are also associated with an increase in caspase 3 activity (Almeida et al., 2013).

Pichardo et al. (2010) revealed that the swim-up procedure in sheep sperm significantly reduces the percentage of spermatozoa expressing caspases 3 and 7, which is accompanied by an increase in chromatin abnormalities in samples with low motility.  They identified that active caspases were located in the implantation fossa region.

Cryopreservation activates the activity of caspases in the sperm of stallions (Ortega-Ferrusola et al., 2008). Caspase-3-positive spermatozoa are immature sperma-tozoa that appear in the ejaculate. Moreover, higher the proportion of such spermatozoa in the stallion’s ejaculate, poorer the cryostability of the samples. Conversely, the lower the activity of caspase 3 and the higher the thiol content, the more cryoprotective the sperm are (Muñoz et al., 2016). Da Silva et al. (2023) proposed to use a multiparametric assessment of stallion sperm using flow cytometry, including the detection of 4-HNE, caspase 3 and 7 activity, live/dead spermatozoa,  and for these purposes it is proposed to investigate live, dead, caspase 3, ROS, mitochondrial membrane potential in stallion semen (Becerro-Rey et al.,  2024).

Protein kinase B maintains the integrity of the mem-branes of ejaculated stallion spermatozoa by inhibiting caspases 3 and 7, preventing the development of apoptosis (Bolaños et al., 2014). At the same time, incubation with rosiglitazone (a drug that preserves the glycolytic activity of spermatozoa) supports the phosphorylation of protein kinase B and reduces the activation of caspase 3, also improves the functioning of the mitochondria of stallion sperm after cryopreservation (Ortiz-Rodriguez et al., 2019). Caselles et al. (2014) demonstrated for the first time the presence of apoptotic bodies in horse semen. The number of apoptotic bodies varied greatly in stallions and positively correlated with caspase 3 activity in fresh samples and negatively with sperm viability and motility after cryopreservation.
 
Caspase 8
 
Caspase 8 (CASP8) is a proteolytic enzyme effector caspase, encoded by the CASP8 gene. The length of the polypeptide chain of the protein is 479 amino acid residues and the molecular weight is 55.391 kDa (Newton et al., 2019).

It is necessary for extrinsic cell death pathways initiated by members of the TNF family. Death receptors (e.g. Fas receptor (CD-95)) recruit DISC after binding of specific TNF family ligands and trimerization. Procaspase-8 is incorporated into this complex by binding through its N-terminal prodomain consisting of two death effector domains to the FADD adapter protein. N-terminal prodomain is joined using a linker of 50 residues to a catalytic domain of 270 amino acids, which contains two sites of cleavage by caspase-8 for autoproteolytic proce-ssing. Within the catalytic domain, there are two additional cleavage sites that allow it to be cleaved into two different subunits, called á or p18 and â or p11. Dimerized procaspase-8 molecules in DISC are activated by reverse cleavage. The activated caspase-8 then initiates a subsequent apoptotic cascade cleaving caspase-3, caspase-7, or Bid. The Bid protein is a link between the external and internal pathways of apoptotic signal transmission (Ghavami et al., 2009; Dyatlova et al., 2018; Keller et al., 2009).

In addition to regulating cell death, caspase-8 also acts as a key mediator of inflammatory cytokine production. After activation of the death receptor or TLR, caspase-8 can stimulate the production of cytokines mediated by NF-kB (TNF–α and IL-6). Caspase-8 can also promote inflammation by regulating the signaling of the NLRP3 inflammasome. This mediates the involvement of caspase 8 in the development of various inflammatory and neurodegenerative diseases (Kumar et al., 2023).
 
Caspase 8 in sperm
 
Caspases 3, 8 and 9 are present in sperm and seminal plasma and are responsible for germ cell apoptosis (Said et al., 2004; Grunewald et al., 2009; Wei et al., 2015; Asadi et al., 2021). In humans, active caspases 8 are localized mainly in the postacrosomal region (Paasch et al., 2004a).  At the same time, it was found that the activity of caspases 8 and 9 in sperm and seminal plasma strongly negatively correlates with sperm concentration and motility (Wei et al., 2015). In oligozoospermia, along with an increase in the activity of caspase 3, there is an increase in the activity of caspase 8, since these enzymes are links of the same signaling pathway. It can be assumed that this condition is associated not only with a violation of meiosis, but also with damage to membranes, which triggers an apoptotic cascade (Almeida et al., 2013).

The possibility of activating the receptor mechanism of apoptosis under the action of cryopreservation is indicated that, after thawing of stallion spermatozoa, exposure of TNR1 and TNFR2 death receptors, as well as the presence of the TNF ligand, was revealed on the plasma membrane. These data were obtained by immunocy-tochemical staining methods (Bolaños et al., 2014). In addition, there is an increase in the expression of IL-1β, TGF-β and corresponding receptors (IL-1R, ACVR1C) in cryopreserved human embryonic stem cells. Activation of the MAP kinase pathway is also detected, which causes activation of caspases. Activation of death receptors may be the result of hyperosmotic shock caused by high concentrations of DMSO, which is used as a cryoprotector (Xu et al., 2010, Ichikawa et al., 2012).

Since the extrinsic pathway of apoptosis can switch to the intrinsic one under certain conditions (Savitskaya and Onishchenko, 2016; Dyatlova et al., 2018). Most researchers detect a simultaneous increase in the activity of caspases in the studied biomaterial. Thus, cryopreser-vation of sperm is accompanied by a significant increase in the activity of caspases 3, 8 and 9 in human sperm (Paasch et al., 2004b; Said et al., 2010). When cryopre-served sperm is thawed from infertile men, the activity of caspases 8 and 9 increases significantly more than in healthy men, indicating lower cryostability (Grunewald et al., 2009). Similar conclusions about the increased activity of caspases 8 and 9 during cryopreservation were reached by Wundrich et al. (2006). In addition, the researchers found that there is a dependence on the concentration of glycerin used during freezing. In the samples where glycerin with a concentration of 14% was used, the increase in caspase activity was more significant than in the samples with 7% glycerin. This is probably due to the toxic effect of glycerol on sperm mitochondria (Wundrich et al., 2006).

Some scientists believe that caspase 8 serves as a switch between apoptosis and necroptosis. Active caspase 8 inhibits the phosphorylation of RIPK1 and RIPK3, which leads to apoptosis. However, when caspase 8 is inactivated, RIPK1 and RIPK3 mutually activate their phosphorylation and subsequently activate the underlying MLKL, causing necroptosis (Li et al., 2021; Hai et al., 2024).
 
Caspase 9
 
The mitochondrial (intrinsic) pathway of apoptosis is triggered by pro-apoptotic BH3 proteins, which can be activated by harmful stimuli, including ROS, gamma radiation and DNA damage. These proteins inhibit the anti-apoptotic Bcl-2, Bcl-xL, thereby weakening the inhibition of the pro-apoptotic factors BAX and BAK. The BAX/BAK-mediated increase in mitochondrial permeability leads to the loss of mitochondrial membrane potential (DψM) and the release of Cytochrome c from the intermembrane space into the cytosol. The release of cytochrome c leads to the activation of caspase 9 through the apoptosome. Activated caspase 9 eventually activates caspase 3, which promotes the cleavage of cytoskeletal proteins and activation of DNases (Chowdhury et al., 2008; Castellini et al., 2021; Dyatlova et al., 2018; Asadi et al., 2021). 

The active form of caspase 9 has a dimeric form. Allosteric inhibition of the enzyme is possible due to phosphorylation and conformational changes. Like other caspases, caspase-9 has three domains: an N-terminal prodomain, a large subunit and a small subunit. The N-terminal prodomain is also called the long prodomain and it contains a caspase activation domain (CARD) motif. The prodomain is linked to the catalytic domain by a linker loop. The linker loop connects the pro domain and the catalytic domain, which consists of large and small subunits. The caspase-9 is activated by dimerization rather than cleavage, although caspase-9 showed full activity in its non-cleaved form, probably due to the long linker loop between the subunits. It is assumed that due to its length and the function of connecting large and small subunits, as well as due to its recruitment and dimerization in the apoptosome, the linker loop moves and gets access to the active site without splitting. Dimerization of caspase-9 leads to rapid autocatalytic cleavage, which results in the formation of caspase-9 (Li et al., 2017; Srinivasula et al., 1996; Renatus et al., 2001).

The non-apoptotic role of caspase-9 includes the regulation of necroptosis, cellular differentiation, innate immune response, maturation of sensory neurons, mitochondrial homeostasis and organization of the cortical-spinal chain. Increased activity of caspase-9 is associated with the progression of amyotrophic lateral sclerosis, retinal detachment, as well as various other neurological, autoimmune and cardiovascular diseases (Avrutsky and Troy, 2021). Caspase 9 is found in all cells and tissues. Caspase 9 in the cell is located in the nucleus, cytoplasm and mitochondria (Zhivotovsky et al., 1999). Its greatest representation is in the tissues of the brain and nervous system, skeletal muscles, liver, pancreas (Srinivasula et al., 1996; Han et al., 2006).
 
Caspase 9 in sperm
 
Caspase 9 is present in human spermatozoa and the mitochondrial fraction of the enzyme is located mainly in the middle piece of the sperm. At the same time, activation of this initiatory caspase most often occurs in sperm endoplasmic reticulum (Paasch et al., 2004a). In immature human spermatozoa, the activity of caspase 9 is initially slightly higher (Zalata et al., 2016). The level of caspase-9 in semen was significantly increased in infertile men compared with healthy fertile subjects from the control group. The activity of casapse-9 spermatozoa has a significant negative correlation with the number of sperma-tozoa, sperm linearity index, sperm motility, sperm velocity, sperm linear velocity and normal morphology (Zalata et al., 2011).

Many researchers have confirmed an increase in the activity of caspases, including 9, with the development of oxidative stress, an increase in the concentration of reactive oxygen species. These factors trigger a cascade of proapoptotic events, which negatively affects the parameters of the spermogram (Lysiak et al., 2007; Mostafa et al., 2016), including in horses (Ball, 2008).

The negative effect of excessive CASP9 activation on spermatogenesis is also confirmed by the fact that the use of its inhibitors improves sperm quality in rats: sperm concentration and motility increase (Zhang et al., 2018).

Cryopreservation/thawing causes activation of caspase 9 in human spermatozoa (Paasch et al., 2004b) and boar spermatozoa (Zeng et al., 2014). The predo-minance of the mitochondrial pathway of apoptosis is also evidenced by a decrease in the level of caspase activity in stallion spermatozoa under the action of bongkrekic acid, a mitochondrial permeability transition pore inhibitor (Ortega et al., 2010), a significant decrease in the level of the mitochondrial membrane potential (ΔψM) in bull spermatozoa (Martin et al., 2004).
 
The use of caspase inhibitors to improve sperm cryopre-servation protocols
 
The caspase family has been actively studied for many years in the development of medicines (Aly et al., 2020; El-Sheref et al., 2020; Poreba et al., 2013). Dhani et al. (2021) described in detail the classification of caspase inhibitors and the possibilities of their use for the treatment of inflammatory, neurodegenerative, metabolic and tumor diseases. Table 1 shows examples of the use of caspase inhibitors 3, 8 and 9 to improve sperm cryopreservation protocols.

Table 1: The use of caspase 3, 8, 9 inhibitors in sperm cryopreservation.



To optimize cryopreservation protocols, substances that inhibit caspase activity, specifically caspases 3, 8 and 9, are utilized not only for their direct effect on these enzymes but also for their ability to indirectly suppress apoptosis. For example, the selective ROCK (Rho-associated protein kinase) inhibitor Y27632 influences the apoptosis cascade by targeting upstream regulatory mechanisms, thereby enhancing cell survival (Table 1). We would also like to focus on the study by Peter et al., (2005), which didn‘t find a positive effect when the pancaspase inhibitor Z-VAD-FMK was added to the stallion sperm in a cryopreservation medium. In this work, only one concentration of Z-VAD-FMK was studied and the sample of the studied stallions was limited to three animals, which indicates the relevance of further studies on the use of caspase inhibitors in cryopreservation of stallion sperm.

A summarizing scheme of caspases 3, 8, 9 participations in sperm apoptosis as application points for caspase inhibitors is shown in Fig 1.

Fig 1: Caspases 3, 8, 9 in sperm apoptosis as application points for inhibitors (Created in BioRender. BioRender.com/y82s086. Agreement number: TT27DL2SEK.).

Apoptosis is one of the well-known mechanisms of sperm quality control. However, increased levels of apoptosis can have an adverse effect on sperm production, ultimately endangering male fertility. Therefore, controlling the rate of apoptosis for male fertility is of particular importance. It follows from the literature data that a higher frequency of apoptosis is observed in the sperm of subfertile men and an assessment of caspase activity is proposed as a diagnostic marker of sperm quality, which provides a more complete sperm analysis for andrological examinations. This aspect is also insufficiently studied in animal husbandry and is quite perspective. It is essential to investigate the activity of caspases both in spermatozoa and in seminal plasma.

In addition, it is relevant to study the types and mechanisms of cell death, as well as the role of caspases in the implementation of these mechanisms in the process of cryopreservation of spermatozoa. This can help in choosing the right cryoprotector, as well as choosing substances that can inhibit apoptosis at different stages, including suppressing the activity of caspases 3,8,9.
The present study was supported by the grant of the Russian Science Foundation No. 20-16-00101-Ð.
 
Disclaimers
 
The views and conclusions expressed in this article are solely those of the authors and do not necessarily represent the views of their affiliated institutions. The authors are responsible for the accuracy and completeness of the information provided, but do not accept any liability for any direct or indirect losses resulting from the use of this content.
The Authors declare that there is no conflict of interest.

  1. Agniswamy, J., Fang, B. and Weber, I. T. (2007). Plasticity of S2- S4 specificity pockets of executioner caspase-7 revea- led by structural and kinetic analysis. The FEBS Journal. 274(18): 4752-4765.

  2. Ahanger, S.A., Wani, U.Y., Rather, M.A., Khan, S.A. and Magray, S.N. (2024). Artificial insemination: A new horizon for scientific goat breeding in kashmir’s backyard farming sector. Bhartiya Krishi Anusandhan Patrika. 39(3): 340-341. doi: 10.18805/ BKAP672.

  3. Almeida, C., Cardoso, M. F., Sousa, M., Viana, P., Gonçalves, A., Silva, J. et al. (2005). Quantitative study of caspase-3 activity in semen and after swim-up preparation in relation to sperm quality. Human Reproduction (Oxford, England). 20(5): 1307-1313. 

  4. Almeida, C., Correia, S., Rocha, E., Alves, A., Ferraz, L., Silva, J. et al. (2013). Caspase signalling pathways in human spermato genesis. Journal of Assisted Reproduction and Genetics. 30(4): 487-495.

  5. Aly, A.A., Sayed, S.M., Abdelhafez, E.S.M., Abdelhafez, S.M.N., Abdel- zaher, W.Y., Raslan, M.A. et al. (2020). New quinoline-2- one/pyrazole derivatives; design, synthesis, molecular docking, anti-apoptotic evaluation and caspase-3 inhibition assay. Bioorganic Chemistry. 94: 103348.

  6. Asadi, A., Ghahremani, R., Abdolmaleki, A. and Rajaei, F. (2021). Role of sperm apoptosis and oxidative stress in male infertility: A narrative review. International Journal of Reproductive Biomedicine. 19(6): 493-504.

  7. Atroschenko, M.M., Zvyagina, V.I., Shitikova, A.M. and Gareski, I.V. (2022). Study of the relationship between the levels of L-camitine and NO metabolites in blood plasma and seminal plasma in stallions. Indian Journal of Animal Research. 56(12): 1483-1487. doi: 10.18805/IJAR.BF-1545.

  8. Atroshchenko, M.M., Bragina, E.E., Zaitsev, A.M., Kalashnikov, V.V., Naumenkova, V.A., Kudlaeva, A.M. et al. (2019). Conser- vation of genetic resources in horse breeding and major structural damages of sperm during semen cryopre- servation in stallions. Nature conservation research. Zapovednaya Nauka. 4(S2): 78-82.

  9. Avrutsky, M.I., Troy, C.M. (2021). Caspase-9:  A multimodal therapeutic target With diverse cellular expression in human disease. Frontiers in Pharmacology. 12: 701301.

  10. Ball, B.A. (2008). Oxidative stress, osmotic stress and apoptosis: Impacts on sperm function and preservation in the horse. Animal Reproduction Science. 107(3-4): 257-267.

  11. Becerro-Rey, L., Martín-Cano, F.E., Sancler-Silva, Y.F.R., Gil, M.C., Ortega-Ferrusola, C. et al. (2024). In vitro, the aging of stallion spermatozoa at 22oC is linked to alteration in Ca2+ and redox homeostasis and may be slowed by regulating metabolism. Theriogenology. 229: 127-137.

  12. Behnam, M., Asadpour, R., Topraggaleh, T. R. and Hamali, H. (2023). Improvement of post-thaw quality and fertilizing ability of bull spermatozoa using Rho kinase inhibitor in freezing extender. Frontiers in Veterinary Science. 10: 1155048.

  13. Boatright, K.M., Salvesen, G.S. (2003). Mechanisms of caspase activation. Current opinion in cell biology. 15(6): 725-731.

  14. Caselles, A.B., Miro Moran, A., Morillo Rodriguez, A., Gallardo Bolanos, J.M., Ortega Ferrusola, C., Salido, G.M. et al. (2014). Identification of apoptotic bodies in equine semen.  Repro- duction in Domestic Animals. 49(2): 254-262. 

  15. Castellini, C., D’Andrea, S., Cordeschi, G., Totaro, M., Parisi, A., Di Emidio, G. et al. (2021). Pathophysiology of mitochondrial dysfunction in human spermatozoa: focus on energetic metabolism, oxidative stress and apoptosis. Antioxidants. 10(5): 695.

  16. Chandra, G., Aggarwal, A., Singh, A.K., Kumar, M., Kushwaha, R., Singh, A. et al. (2011). Negative energy balance and reproduction: A review. Agricultural Reviews. 32(4): 246-254. 

  17. Chowdhury, I., Tharakan, B. and Bhat, G.K. (2008). Caspases-an update. Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology. 151(1): 10-27.

  18. Contreras, M.J., Arias, M.E., Fuentes, F., Muñoz, E., Bernecic, N., Fair, S. et al. (2023). Cellular and molecular consequences of stallion sperm cryopreservation: Recent approaches to improve sperm survival. Journal of Equine Veterinary Science. 126: 104499.

  19. Crowley, L.C., Waterhouse, N.J. (2016). Detecting Cleaved Caspase-3 in Apoptotic Cells by Flow Cytometry. Cold Spring Harbor Protocols. 2016(11): 10.1101/pdb.prot087312.

  20. Da Silva, C.M.B., Cano, F.E.M., Gaitskell Phillips, G. and Vega, F.J.P. (2023). Multiparametric flow cytometry for determination of viability, caspase 3 and 7 activity and lipid peroxidation adduct (4 Hydroxynonenal) in equine spermatozoa. Current Protocols. 3(9): e885.

  21. Dhani, S., Zhao, Y. and Zhivotovsky, B. (2021). A long way to go: caspase inhibitors in clinical use. Cell Death and Disease. 12(10):  949.

  22. Dyatlova, A.S., Dudov, A.V., Linkova, N.S. and Khavinson, V.Kh. (2018). Molecular markers of caspase-dependent and mitochondrial apoptosis: The role of pathology and cell senescence. Biology Bulletin Reviews. 138 (2): 126-137. 

  23. El-Sheref, E.M., Aly, A.A., Alshammari, M.B., Brown, A.B., Abdel- Hafez, S.M.N., Abdelzaher, W.Y. et al. (2020). Design, synthesis, molecular docking, antiapoptotic and caspase-3 inhibition of new 1, 2, 3-triazole/bis-2 (1 H)-quinolinone hybrids. Molecules. 25(21): 5057.

  24. Fomina, M.A., Kudlaeva, A.M. (2016). In vitro studies of L-carnitine action on lysosomal cysteine proteolysis alone and in oxidative stress. Nauka Molodykh (Eruditio Juvenum). 1: 55-59. 

  25. Fomina, M.A., Kudlaeva, A.M. and Ryabkov, A.N. (2017). In vitro effect of L-carnitine on the activity of lysosomal cysteine proteases and the state of lysosomal membrane. I.P. Pavlov Russian Medical Biological Herald. 25(1): 14-20.

  26. Bolaños, J.G., Da Silva, C.B., Munoz, P.M., Rodriguez, A.M., Davila, M.P., Rodriguez-Martinez, H. et al. (2014). Phosphory- lated AKT preserves stallion sperm viability and motility by inhibiting caspases 3 and 7. Reproduction. 148(2): 221-35.

  27. Ghavami, S., Hashemi, M. ande, S.R., Yeganeh, B., Xiao, W., Eshraghi, M. et al. (2009). Apoptosis and cancer: mutations within caspase genes. Journal of medical genetics. 46(8): 497-510.

  28. Grunewald, S., Sharma, R., Paasch, U., Glander, H.J. and Agarwal, A. (2009). Impact of caspase activation in human spermatozoa. Microscopy Research and Technique. 72(11): 878-888.

  29. Hai, E., Li, B., Zhang, J. and Zhang, J. (2024). Sperm freezing damage: The role of regulated cell death. Cell Death Discovery. 10(1): 239.

  30. Han, Y., Chen, Y.S., Liu, Z., Bodyak, N., Rigor, D., Bisping, E. et al. (2006). Sperm freezing damage: The role of regulated cell death. Cell Death Discovery Overexpression of HAX- 1 protects cardiac myocytes from apoptosis through caspase-9 inhibition. Circulation Research. 99(4): 415-423.

  31. Ichikawa, H., Nakata, N., Abo, Y., Shirasawa, S.,Yokoyama, T., Yoshie, S. et al. (2012). Gene pathway analysis of the mechanism by which the Rho-associated kinase inhibitor Y-27632 inhibits apoptosis in isolated thawed human embryonic stem cells. Cryobiology. 64: 12-s22.

  32. Keller, N., Mareš, J., Zerbe, O. and Grütter, M.G. (2009). Structural and biochemical studies on procaspase-8: New insights on initiator caspase activation. Structure. 17(3): 438-448.

  33. Kesavardhana, S., Malireddi, R. and Kanneganti, T. D. (2020). Caspases in Cell Death, Inflammation and Pyroptosis. Annual Review of Immunology. 38: 567-595.

  34. Kumar, S., Budhathoki, S., Oliveira, C.B., Kahle, A.D., Calhan, O.Y., Lukens, J.R. et al. (2023). Role of the caspase-8/RIPK3 axis in Alzheimer’s disease pathogenesis and Aâ-induced NLRP3 inflammasome activation. JCI insight. 8(3): e157433. 

  35. Lavrik, I.N., Golks, A. and Krammer, P.H. (2005). Caspases: Pharma- cological manipulation of cell death. The Journal of clinical investigation. 115(10): 2665-2672.

  36. Li, X., Zhong, C.Q., Wu, R., Xu, X., Yang, Z.H., Cai, S. et al. (2021). RIP1- dependent linear and nonlinear recruitments of caspase-8 and RIP3 respectively to necrosome specify distinct cell death outcomes. Protein and Cell. 12(11): 858-876.

  37. Li, P., Nijhawan, D. and Wang, X. (2004). Mitochondrial activation of apoptosis. Cell. 116(2 Suppl): S57-S59. 

  38. Li, P., Zhou, L., Zhao, T., Liu, X., Zhang, P., Liu, Y., Zheng, X. et al. (2017). Caspase-9: structure, mechanisms and clinical application. Oncotarget. 8(14): 23996-24008.

  39. Lysiak, J.J., Zheng, S., Woodson, R. and Turner, T.T. (2007). Caspase-9- dependent pathway to murine germ cell apoptosis: Mediation by oxidative stress, BAX and caspase 2. Cell and tissue research. 328(2): 411-419.

  40. Madkar, A.R., Boro, P. and Abdullah, M. (2022). Estrus detection methods in dairy animals-advances and the prospects: A review. Agricultural Reviews. 43(3): 362-367.

  41. Man, S.M., Karki, R. and Kanneganti, T. D. (2017). Molecular mecha- nisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunological Reviews. 277(1): 61-75.

  42. Marchetti, C., Gallego, M.A., Defossez, A., Formstecher, P. and Marchetti, P. (2004). Staining of human sperm with fluorochrome labeled inhibitor of caspases to detect activated caspases: Correlation with apoptosis and sperm parameters. Human Reproduction. 19(5): 1127-1134.

  43. Martin, G., Sabido, O., Durand, P. and Levy, R. (2004). Cryopre- servation induces an apoptosis-like mechanism in bull sperm. Biology of Reproduction. 71(1): 28-37.

  44. Moreno, R.D., Alvarado, C.P. (2006). The mammalian acrosome as a secretory lysosome: New and old evidence. Molecular Reproduction and Development. 73(11): 1430-1434. 

  45. Mostafa, T., Rashed, L. and Taymour, M. (2016). Seminal cyclooxy- genase relationship with oxidative stress in infertile oligoas thenoteratozoospermic men with varicocele. Andrologia. 48(2): 137-142.

  46. Muñoz, P.M., Ferrusola, C.O., Lopez, L.A., Del Petre, C., Garcia, M.A., de Paz Cabello, P., Anel, L. and Peña, F.J. (2016). Caspase 3 activity and lipoperoxidative status in raw semen predict the outcome of cryopreservation of stallion spermatozoa.  Biology of Reproduction. 95(3): 53. 

  47. Murphy, C.J., Richburg, J.H. (2014). Implications of Sertoli cell induced germ cell apoptosis to testicular pathology. Spermatogenesis.  4(2): e979110.

  48. Newton, K., Wickliffe, K.E., Maltzman, A., Dugger, D.L., Reja, R., Zhang, Y., et al. (2019). Activity of caspase-8 determines plasticity between cell death pathways. Nature. 575(7784): 679-682.

  49. Ortega Ferrusola, C., Gonzalez Fernandez, L., Salazar Sandoval, C., Macias Garcia, B., Rodriguez Martinez, H., Tapia, J.A. et al. (2010). Inhibition of the mitochondrial permeability transition pore reduces «apoptosis like» changes during cryopreser- vation of stallion spermatozoa. Theriogenology. 74: 458-465.

  50. Ortega Ferrusola, C., Sotillo Galan, Y., Varela Fernandez, E., Gallardo Bolanos, J.M., Muriel, A., Gonzalez Fernandez, L. et al. (2008). Detection of “apoptosis like” changes during the cryopreser- vation process in equine sperm. Journal of Andrology. 29(2): 213-221.

  51. Ortiz-Rodriguez, J.M., Balao da Silva, C., Masot, J., Redondo, E., Gazquez, A., Tapia, J.A. et al. (2019). Rosiglitazone in the thawing medium improves mitochondrial function in stallion sperma- tozoa through regulating Akt phosphorylation and reduction of caspase 3. PLoS One. 14(7): e0211994.

  52. Paasch, U., Grunewald, S., Agarwal, A. and Glandera, H.J. (2004a). Activation pattern of caspases in human spermatozoa. Fertility and Sterility. 81(1): 802-809.

  53. Paasch, U., Sharma, R.K., Gupta, A.K., Grunewald, S., Mascha, E.J., Thomas, A.J. et al. (2004b). Cryopreservation and thawing is associated with varying extent of activation of apoptotic machinery in subsets of ejaculated human spermatozoa. Biology of Reproduction. 71(6): 1828-1837.

  54. Pagano, N., Longobardi, V., De Canditiis, C., Zuchegna, C., Romano, A., Michal Andrzej, K. et al. (2020). Effect of caspase inhibitor Z VAD FMK on bovine sperm cryotolerance.  Reproduction in Domestic Animals. 55(4): 530-536.

  55. Peter, A.T., Colenbrander, B. and Gadella, B.M. (2005). Effect of caspase inhibitors on the post-thaw motility and integrity of acrosome and plasma membrane of cryopreserved equine spermatozoa. Indian Journal of Experimental Biology. 43(6): 483. 

  56. Pichardo, A.I., Aragón-Martínez, A., Ayala-Escobar, M.E. and Domínguez- Vara, I.A. (2010). Viability tests, active caspase-3 and - 7 and chromatin structure in ram sperm selected using the swim-up procedure. Journal of Andrology. 31(2): 169-176.

  57. Poreba, M., Strózyk, A., Salvesen, G. S. and Drag, M. (2013). Caspase substrates and inhibitors. Cold Spring Harbor Perspectives in Biology. 5(8): a008680.

  58. Renatus, M., Stennicke, H.R., Scott, F.L., Liddington, R.C. and Salvesen, G.S. (2001). Dimer formation drives the activation of the cell death protease caspase 9. Proceedings of the National Academy of Sciences of the United States of America. 98(25): 14250-14255.

  59. Said, T.M., Gaglani, A. and Agarwal, A. (2010). Implication of apoptosis in sperm cryoinjury. Reproductive biomedicine online. 21(4): 456-462. 

  60. Said, T.M., Paasch, U., Glander, H.J. and Agarwal, A. (2004). Role of caspases in male infertility. Human reproduction update. 10(1): 39-51.

  61. Savitskaya, M.A., Onishchenko, G.E. (2016). Apoptosis in cryo- preserved eukaryotic cells. Biochemistry (Moscow). 81(5): 445-452.

  62. Shalini, S., Dorstyn, L., Dawar, S. and Kumar, S. (2015). Old, new and emerging functions of caspases. Cell Death and Differentiation. 22(4): 526-539. 

  63. Sharma, M.K. (2015). Effects of cooling rate alterations in critical temperature range and supplementation of anti apoptotic proteins on cryosurvival of Buffalo bull sperm (Doctoral dissertation, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana). 

  64. Shitikova, A.M., Atroshchenko, M.M. and Zvyagina, V.I. (2023). Thiol Cathepsins and Oxidative Modification of Stallion’s Seminal Plasma Proteins with Normal and Low Percentage of Live Spermatozoa Post Cryopreservation. Indian Journal of Animal Research. 58(10): 1658-1664. doi: 10.18805/ IJAR.BF-1672.

  65. Shitikova, A.M., Atroshchenko, M.M., Zvyagina, V.I., Belskikh, E.S. and Romanova, A.I. (2024). Importance of L-carnitine as bioche- mical marker for semen quality preservation after cryopre- servation in stallions. Indian Journal of Animal Research. doi: 10.18805/IJAR.BF-1792.

  66. Srinivasula, S.M., Fernandes-Alnemri, T., Zangrilli, J., Robertson, N., Armstrong, R.C., Wang, L. et al. (1996). The Ced-3/ interleukin 1beta converting enzyme-like homolog Mch6 and the lamin-cleaving enzyme Mch2alpha are substrates for the apoptotic mediator CPP32. The Journal of Biological Chemistry. 271(43): 27099-27106.

  67. Stennicke, H.R., Salvesen, G.S. (1997). Biochemical characteristics of caspases-3, -6, -7 and -8. The Journal of Biological Chemistry. 272(41): 25719-25723.

  68. Upadhyay, V.R., Ramesh, V., Dewry, R.K., Kumar, G., Raval, K. and Patoliya, P. (2021). Implications of cryopreservation on structural and functional attributes of bovine spermatozoa: An overview. Andrologia. 53(8): e14154.

  69. Van Opdenbosch, N., Lamkanfi, M. (2019). Caspases in Cell Death, Inflammation and Disease. Immunity. 50(6): 1352-1364. 

  70. Walters, J., Pop, C., Scott, F. L., Drag, M., Swartz, P., Mattos, C. et al. (2009). A constitutively active and uninhibitable caspase- 3 zymogen efficiently induces apoptosis. The Biochemical Journal. 424(3): 335-345. 

  71. Wang, X., Sharma, R.K., Sikka, S.C., Thomas Jr, A.J., Falcone, T. and Agarwal, A. (2003). Oxidative stress is associated with increased apoptosis leading to spermatozoa DNA damage in patients with male factor infertility. Fertility and sterility.  80(3): 531-535. 

  72. Wei, X., Li, Q., Han, Z., Lin, D. and Yu, P. (2015). Differences in caspase-8 and -9 activity and sperm motility in infertile males of Li nationality in China. International journal of clinical and experimental medicine. 8(3): 4721-4726.

  73. Weng, S.L., Taylor, S.L., Morshedi, M., Schuffner, A., Duran, E.H., Beebe, S. et al. (2002). Caspase activity and apoptotic markers in ejaculated human sperm. Molecular human reproduction. 8(11): 984-991. 

  74. Wundrich, K., Paasch, U., Leicht, M. and Glander, H.J. (2006). Activation of caspases in human spermatozoa during cryopreservation -an immunoblot study. Cell and tissue banking. 7(2): 81-90. 

  75. Wyllie A. H. (1997). Apoptosis: an overview. British medical bulletin. 53(3): 451-465. 

  76. Xu, X., Cowley, S., Flaim, C.J., James, W., Seymour, L. and Cui, Z. (2010). The roles of apoptotic pathways in the low recovery rate after cryopreservation of dissociated human embryonic stem cells. Biotechnology progress. 26(3): 827-837.

  77. Xu, Y. W., Ou, N.J., Song, Y.X., Wang, X.H., Kang, J.Q., Yang, Y.J. et al. (2020). Seminal plasma miR-210-3p induces spermato- genic cell apoptosis by activating caspase-3 in patients with varicocele. Asian journal of andrology. 22(5): 513-518. 

  78. Zalata, A.A., Mokhtar, N., Atwa, A., Khaled, M. and Shaker, O.G. (2016). The Role of Protamine 2 Gene Expression and Caspase 9 Activity in Male Infertility. The Journal of urology. 195(3): 796-800.

  79. Zalata, A., El-Mogy, M., Abdel-Khabir, A., El-Bayoumy, Y., El-Baz, M. and Mostafa, T. (2011). Sperm caspase-9 in oligoasthe noteratozoospermic men with and without varicocele. Fertility and sterility. 96(5): 1097-1099. 

  80. Zeng, C., Tang, K., He, L., Peng, W., Ding, L., Fang, D. et al. (2014) Effects of glycerol on apoptotic signaling pathways during boar spermatozoa cryopreservation. Cryobiology. 68: 395-404. 

  81. Zhang, K., Ge, Z., Fu, L., An, Q., Zhou, F., Guo, Y. et al. (2018). Qilin pills alleviate oligoasthenospermia by inhibiting Bax- caspase-9 apoptosis pathway in the testes of model rats. Oncotarget. 9(31): 21770-21782.

  82. Zhang, M.H., Zhang, A.D., Shi, Z.D., Wang, L.G. and Qiu, Y. (2015). Changes in levels of seminal nitric oxide synthase, macrophage migration inhibitory factor, sperm DNA integrity and Caspase-3 in Fertile Men after Scrotal Heat Stress. PloS one. 10(10): e0141320. 

  83. Zhivotovsky, B., Samali, A., Gahm, A. and Orrenius, S. (1999). Caspases: Their intracellular localization and translocation during apoptosis. Cell death and differentiation. 6(7): 644-651. 

Editorial Board

View all (0)