Open access peer-reviewed chapter

Endoplasmic Reticulum Stress during Mammalian Follicular Atresia

Written By

Nayeli Torres-Ramírez, Rosario Ortiz-Hernández, M. Luisa Escobar-Sánchez, Olga M. Echeverría-Martínez and Gerardo H. Vázquez-Nin

Submitted: 25 September 2018 Reviewed: 23 November 2018 Published: 15 February 2019

DOI: 10.5772/intechopen.82687

From the Edited Volume

Endoplasmic Reticulum

Edited by Angel Català

Chapter metrics overview

1,349 Chapter Downloads

View Full Metrics

Abstract

Follicles are ovarian structures that contain a single germ cell. During the mammalian reproductive lifetime, ovarian follicles mature through the process of follicular development, with the aim of selecting oocytes for ovulation. As part of this process, several follicles are eliminated by means of follicular atresia, a mechanism that mainly involves apoptosis. Nevertheless, it has been shown that there are other routes of programmed cell death in the ovary including autophagy, paraptosis, and necroptosis. Surprisingly, the endoplasmic reticulum is involved in these different programmed cell death pathways. Moreover, there are several evidences for the pathways triggered by intra- and extracellular signals in endoplasmic reticulum-induced cell death. Thus, it is important to analyze the participation of endoplasmic reticulum in follicular atresia.

Keywords

  • ovary
  • follicular atresia
  • endoplasmic reticulum
  • apoptosis
  • autophagy
  • paraptosis
  • necroptosis

1. Introduction

The endoplasmic reticulum plays several important roles in normal cellular physiology. Some functions include protein synthesis, folding, and distribution to the Golgi apparatus. Alterations in protein synthesis inside the endoplasmic reticulum have been related to the trigger of different programmed cell death routes such as necroptosis, apoptosis, autophagy, and paraptosis, with apoptosis being the most studied process.

The mammalian ovary is an excellent model to study the mechanisms of programmed cell death because 99% of the follicles, the functional units of the ovary, undergo degeneration through follicular atresia, which maintains intraovarian homeostasis. Follicular atresia involves the physiological elimination of most germinal cells (oocytes) before they are ovulated, both in fetal and reproductive lives.

The presence of different programmed cell death pathways in follicular atresia have recently been shown, and these can be directly related to endoplasmic reticulum signaling. In this chapter we describe evidences of the linkage between endoplasmic reticulum alterations and programmed cell death, with special emphasis on follicular atresia.

Advertisement

2. Follicular development and atresia

The mammalian ovary is a paired organ that is responsible for generating competent oocytes for successful fertilization and early embryonic development. To do this, these germinal cells need to mature within transient functional complexes called follicles. Follicles form for an oocyte surrounded by somatic cells. During reproductive life, follicles are continuously recruited into the pool of growing follicles and change their size, morphology, and physiology, leading to different stage classifications including primordial, primary, secondary, and antral (Figure 1).

Figure 1.

Ovary of mouse. Follicles are in different stages of growth. Primordial (P), primary (head arrow), secondary (asterisk), and antral (A) follicles.

At birth, the ovaries contain a fixed number of nongrowing primordial follicles, characterized by an oocyte enclosed by flattened pre-granulosa cells. In primary follicles, the oocyte is surrounded by a monolayer of cubical granulosa cells. Secondary follicles are formed by two or more layers of granulosa cells. Antral follicles accumulate fluid and develop an antral cavity. The accumulation of fluid is useful for transporting nutrients and waste products.

Follicular growth is a continuous process that is under strict control by hormones, growth factors, cytokines, and environmental factors. Follicle-stimulating hormone (FSH), luteinizing hormone (LH), insulin-like growth factor (IGF)-I, and estradiol are the principal regulators of follicular growth. FSH, a gonadotropin secreted by the pituitary gland, together with estradiol and IGF-I, is responsible for stimulating follicular growth and maturation. Moreover, FSH, LH, and estradiol enhance IGF-I secretion [1]. Additionally, FSH stimulates granulosa cells to develop LH receptor sites. The main function of LH is stimulating ovulation.

Several follicles grow and undergo ovulation, releasing an oocyte that is available for fertilization, but the principal destiny of ovarian follicles is follicular atresia, which is a physiological process that eliminates more than 99% of the follicles. Follicular atresia can occur in all stages of follicular development and ensures that only healthy follicles that contain optimal quality oocytes will be ovulated. Follicular degeneration occurs by programmed cell death (PCD). Apoptosis is the main route of follicular atresia, but may not be the only process involved (Figure 2). Other forms of PCD such as autophagy and paraptosis may also participate in this process [2, 3, 4].

Figure 2.

Transmission electron microscope images of granulosa cells in different programmed cell death pathways. (a) healthy granulosa cell, (b) apoptotic body with highly condensed chromatin (cc), (c) autophagic cell with autophagic vesicles (head arrow), and (d) paraptotic granulosa cell with endoplasmic reticulum swelling (asterisk). Bars (a–c) 500 nm, and (d) 2 μm.

Advertisement

3. The endoplasmic reticulum and cell death

The endoplasmic reticulum (ER) is the organelle that is responsible for the folding and maturation of both transmembrane proteins and proteins that follow the route of secretion. Protein folding is facilitated by chaperones and oxidoreductases including binding immunoglobulin protein/glucose-regulated protein 78-kDa (BiP/GRP78), calnexin, calreticulin, and protein disulfide isomerase (PDI). An increase of cellular translational activity is possible under both normal and altered conditions, causing an overload of accumulating misfolding or unfolded proteins inside the ER. During ER stress, damaged proteins need to be degraded, but there is a limited number of proteases in the ER, and thus misfolded proteins are ejected from the ER and returned to the cytoplasm to be ubiquitinated and degraded by the 26S proteasome. These events are collectively referred to as ER-associated degradation (ERAD) [5]. Also, ER stress triggers the unfolded protein response (UPR), which is orchestrated by three ER-resident UPR sensors, inositol-requiring kinase 1 (IRE1), protein kinase R-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6) [6, 7].

The UPR establishes an adaptive program aimed at re-establishing ER homeostasis by increasing the folding capacity of the cell, reducing protein synthesis, and enhancing the clearance of abnormally folded proteins and damaged organelles.

The proteins PERK and IRE1α and β are important players during UPR because they undergo oligomerization and autophosphorylation due to their interactions with peptides and unfolded proteins [8, 9]. Additionally, IRE1 promotes the unconventional splicing of X-box binding protein 1 (XBP-1) mRNA and an unspecific decrease of mRNAs better known as regulated IRE1-dependent decay (RIDD) [10, 11]. Afterward, the protein XBP-1 is translocated to the nucleus to activate the transcription of chaperones and ERAD factors [12]. RIDD suppresses protein inflow by degrading the mRNA of proteins with signal peptides or proteins with transmembrane domains, and in this manner RIDD permits proteins that are incorrectly folded inside the ER to be folded correctly [10]. PERK phosphorylates eukaryotic translation initiation factor (eIF2a), which then accumulates on the cytosolic side and leads to the downregulation of translation and enhances the translation of Grp78 and the transcription factor ATF4 [13, 14]. It has been shown that during early mouse embryonic development, Grp78 suppresses ER stress and pro-apoptotic pathways via ER signaling [15]. ATF6 is regulated by proteolysis in the Golgi apparatus, allowing the N-terminal fragments to be translocated into the nucleus where they function as a transcription factor [16, 17]. The processing of both ATF6- and IRE1α-mediated splicing of XBP1 mRNA is required for the full activation of the UPR [18].

UPR works like a protection mechanism. For example, in pancreatic beta cell line INS-1E, glucosamine and high glucose induce UPR activation and generate a feedback loop at the level of insulin transcription [19]. However, chronic or irreversible UPR can trigger cell death pathways, mainly apoptosis, but ER stress can induce other programmed cell death mechanisms including autophagy, necroptosis, and paraptosis.

3.1 The ER and follicular atresia

Morphological ER disturbances during follicular atresia have been observed for a long time. Henderson et al. [20] observed a higher surface area of endoplasmic reticulum in granulosa cells cultured from atretic follicles. Moreover, researchers have used electron microscopy to observe the dilation and disintegration of RER cisterns and the swelling of mitochondria [21].

These morphological disturbances in ovaries are associated with ER stress and UPR activation under both physiological and pathological conditions [22]. UPR is present during follicular growth and maturation and follicular atresia and in the corpus luteum. ER stress during follicular growth and maturation has been evidenced by means of the expression of XPB1 and heat shock 70 kDa protein 5 (HSPA5) accompanied by the activation of IRE1 and PERK [23]. The ER stress level and cellular response depend on the signal and its intensity. It has been shown that a lipid-rich intrafollicular environment induces ER stress and impaired oocyte nuclear maturation [24]. Likewise, in the ovary a moderate activation of ER stress depends upon PERK and p38 signaling [25], evidencing a UPR response in the cells of this organ.

Advertisement

4. Apoptosis

Apoptosis, the term proposed by Kerr et al. [26], describes an intrinsic suicide mechanism that involves cell shrinkage and the loss of cell contacts, chromatin condensation, and cleavage [27]. This process is better known as programmed cell death type 1 (PCD type 1). The biochemical activation of apoptosis can be directed through extrinsic and intrinsic pathways. The extrinsic pathway is initiated by the activation of cell surface death receptors to their ligands, like the Fas Ligand and TNF. After binding, apoptotic signals are transmitted through dead effector domains and caspase recruitment domains. The intrinsic pathway is governed by a variety of cellular stresses including DNA damage, endoplasmic reticulum stress, and nutrient deprivation, which culminates in mitochondrial outer membrane permeabilization (MOMP), resulting in the release of mitochondrial proteins including cytochrome c and Smac/DIABLO. Apoptosis pathways converge on a common machinery of cell destruction that is activated by caspases, a family of cysteine proteases that cleave after an aspartate residue [28, 29]. The caspases implicated in apoptosis are divided into initiators and executioners, where initiator caspases (caspase-8 and caspase-9) activate the executor caspases (caspase-3, caspase-6, and caspase-7).

The Bcl-2 family, which are central regulators of MOMP, are a large class of both pro- and anti-apoptotic proteins. The Bcl-2 family is divided into three subfamilies: multidomain anti-apoptotic such as BCL-2, BCL-XL (BCL2L1), MCL-1, BCL-W (BCL2L2), and A1 (BCL2A1), multidomain pro-apoptotic such as BAX and BAK, and pro-apoptotic BH3-only molecules that include BID, BIM, PUMA (p53 upregulated modulator of apoptosis), and NOXA [30]. BH3-only proteins antagonize anti-apoptotic BCL-2 proteins to release and activate Bak/Bax [31]. Bax and Bak induce external membrane mitochondrial permeabilization and cytochrome c release [32]. Nevertheless, some death stimuli can trigger caspase-independent cell death pathways where other organelles such as the endoplasmic reticulum and the mitochondria have an important function in the release and activation of death factors [33].

In atretic follicles, this PCD was thoroughly described by Tilly et al. [34] and can be conducted through the intrinsic or the extrinsic pathway [35]. In ovaries, apoptosis can be triggered by deprivation of various signal molecules, survival factors, growth factors (IGF and EGF), and gonadotropins (FSH and LH). Apoptosis can occur in both oocytes and somatic cells. Cell elimination has been observed in follicles in different stages of development, from fetal to adult organisms [3, 36, 37, 38]. Although different routes of PCD can occur during follicular atresia, apoptosis plays a major role (Figure 2b).

4.1 The role of the ER in apoptosis

Apoptosis is triggered by chronic or irreversible ER stress and UPR and occurs through either the extrinsic or intrinsic pathway. Further, apoptosis can be carried out by two pathways, a classical Bax-/Bak-dependent apoptotic response that can be inhibited by ERK1/2 signaling and an alternative ERK1-/2- and Bax-/Bak-independent pathway [39]. No single component is entirely necessary, but the interaction of many different mechanisms results in apoptosis during ER stress [40]. Under ER stress Bax and Bak interact with the cytosolic region of IRE1α, which is required for the modulation of IRE1α signaling [41].

The activity of the BH3-only protein Bim is induced through different pathways. The first one involves protein phosphatase 2A-mediated dephosphorylation, which prevents its ubiquitination and the proteasomal degradation of Bim. A second pathway is direct transcriptional induction that is C/EBP homologous protein (CHOP)-C/EBPalpha-mediated, and a third comprises a repression of miRNAs led by PERK [42, 43]. On the other hand, PUMA, p53, and NOXA contribute to ER stress-induced apoptosis [44].

It has been reported that CHOP (a transcription factor of pro-apoptotic proteins such as Bim) increases during ER stress [45]. ATF4 and CHOP increase a generalized protein synthesis, provoking ATP depletion, oxidative stress, and cell death [46]. Also, IRE1α degrades the miRNA that represses caspase-2 mRNA translation, which causes an increase in the protein levels of this initiator protease of the mitochondrial apoptotic pathway [47].

4.2 The role of the ER in apoptosis during follicular atresia

ER stress and UPR during follicular atresia are not fully understood; however, there are several evidences of these processes in the ovary. For example, cisplatin, a widely used chemotherapeutic agent, can induce ER stress, which promotes apoptosis and autophagy in granulosa cells, causing excessive follicle loss and endocrine disorders [48].

In goat ovaries, ER stress is involved in follicular atresia through ATF6 and PERK/eIF2α/ATF4 signaling. Furthermore CHOP, caspase-12, and Grp78 proteins are upregulated in apoptotic granulosa cells during follicular atresia [49, 50]. ATF6 is a protein that is extensively distributed in the granulosa cells of ovarian follicles and oocytes in adult mice, and the amount of ATF6 increases in the presence of FSH and LH. ATF6 regulates apoptosis, the cell cycle, steroid hormone synthesis, and other modulators of folliculogenesis in granulosa cells, which may impact the development, ovulation, and atresia of ovarian follicles [51].

The presence of apoptosis-inducing factor (AIF) has been identified in granulosa cells. This protein mediates caspase-independent apoptosis and causes chromatin condensation and DNA fragmentation. AIF expression increases during follicular atresia, and AIF depletion protects ER stress-mediated goat granulosa cell apoptosis [52].

Reactive oxygen species (ROS) generation and oxidative stress can be upstream or downstream UPR targets. That is, UPR is interconnected with different enzymatic mechanisms of ROS generation, and they may depend on Ca2+ levels, ROS themselves, and PDI, which associates with NADPH oxidase and regulates its function [53]. ROS are pro-apoptotic factors in antral follicles. During oxidative stress, JNK activates FoxO1, which increases PUMA and induces apoptosis in granulosa cells [54]. Furthermore, pentosidine, a biomarker for advanced glycation end products, is accumulated in apoptotic human oocytes and increases with age [55].

UPR and ER stresses also have important roles in the regulation of corpus luteum (CL) regression. The overexpression of p-JNK, CHOP, caspase-12, and active caspase-3 during CL regression points to ER stress-dependent apoptosis [56, 57].

Advertisement

5. Autophagy

Autophagy is a catabolic pathway of cell constituents that contributes to cell survival in response to stress. Autophagy does not cause a loss of cell chemical components because the cell reutilizes them. There are three major types of autophagy, microautophagy, chaperon-mediated autophagy, and macroautophagy.

In microautophagy, vesicles bud into the lysosomal lumen by direct invagination of the boundary membrane, resulting in degradation of both cytoplasmic components and the lysosomal membrane by lysosomal hydrolases. This process involves sequential stages of vacuole invagination and vesicle scission [58].

Chaperon-mediated autophagy is the selective transport of proteins into lysosomes. The first step is protein recognition and lysosomal targeting. Protein recognition takes place in the cytosol through the binding of hsc70 to a KFERQ-like motif present in all chaperon-mediated autophagy substrates [59]. In the second step, proteins bind to receptors at the lysosomal membrane, Lamp2A, or a similar protein receptor for subsequent translocation and lysosomal degradation [60]. Receptors are subcompartmentalized in lipid microdomains to engage the processes of degradation, multimerization, and membrane retrieval [61].

Macroautophagy, also referred to as autophagy, involves the engulfment of cytoplasmic portions in a nonselective manner, as well as the degradation of specific proteins, organelles, and invading bacteria by a selective autophagy. Autophagy begins with the formation of an isolation membrane, the phagophore, which is a disk-like structure where the Atg machinery assembles. An isolation membrane grows to generate a double-membrane autophagosome, followed by elongation to form a mature autophagosome that captures cytosolic cargo. The fusion of mature autophagosomes with endosomes or lysosomes results in a single-membrane autolysosome where cargo is degraded by acid hydrolases [62].

Autophagy (Atg)-related proteins are the core machinery for autophagosome biogenesis and consist of several functional units: the ULK1-Atg13-FIP200-Atg101 protein kinase complex; the PI3K class III complex containing the core proteins VPS34, VPS15, and beclin 1; the PI3P-binding WIPI/Atg18-Atg2 complex; Atg9A; and the ubiquitin-like Atg5/Atg12 and Atg8/LC3 conjugation systems [63].

Autophagosome maturation involves the clearance of PI3P by Ymr1, a PI3P phosphatase, triggering the dissociation of the Atg machinery. Mature autophagosomes are transported to lysosomes through the microtubule cytoskeleton. The FYVE and coiled-coil domain containing 1 (FYCO1) protein binds to LC3, PI3P, and the small GTPase Rab7 and acts as an adaptor between autophagosomes and microtubules [64, 65]. Finally, the autolysosome is generated by autophagosome and lysosome fusion, where sequestered cargos are digested.

5.1 The role of the ER in autophagy

Autophagy and ER stress can be physiological processes in organisms. For example, they regulate endometrial function by modulating the mTOR pathway [66]. Also, autophagy contributes to the recovery of cell homeostasis after ER stress. During ER stress, damaged proteins are degraded by ERAD. However, some misfolded proteins are resistant, so autophagy is a final cell protection strategy deployed against ER-accumulated cytotoxic aggregates that cannot be removed by ERAD [67]. Additionally, ubiquitin is a common signal for both the ubiquitin-proteasome system and autophagy. In the mouse neuroblastoma cell line neuro-2a treated with tunicamycin, an ER stress inductor, the proteins involved in proteasomal degradation were downregulated, while proteins involved in ubiquitination were upregulated. Moreover, tunicamycin triggered autophagy, suggesting that it may serve as a compensatory effect to proteasomal degradation [68]. Also, ER-resident chaperones and enzymes that reduce the overload of misfolded proteins need to be removed by autophagy.

The structure or phagophore assembly site (PAS) localizes proximal to the ER. Autophagosome formation and transport to the vacuole are stimulated in an Atg protein-dependent manner. ER stress can induce an autophagic response because it increases Atg1 kinase activity and reflects both the nutritional status and autophagic state of the cell [69]. ER exit sites are essential for autophagy and are proximal to the PAS. Sec62, a constituent of the translocon complex that regulates protein import into the mammalian ER, intervenes during recovery from ER stress to selectively deliver ER components to the autolysosomal system for clearance and therefore is a critical molecular component in the maintenance and recovery of ER homeostasis [70].

The eIF2α/ATF4 pathway directs an autophagy gene transcriptional program in response to amino acid starvation or ER stress. The eIF2α kinase and the transcriptional factors ATF4 and CHOP are required to increase the transcription of a set of genes implicated in the formation, elongation, and function of the autophagosome, including Atgs and beclin 1, increasing the capacity to maintain autophagy in stressed cells. These autophagy genes exhibit different dependencies on ATF4 and CHOP, which means that they have a differential transcriptional response according to the stress intensity [71]. In human heart failure, the overexpression of the ER stress markers Grp78, PERK, CHOP, and ATF3 correlates with the expression of autophagy genes [72].

IRE1, a UPR sensor, has two isoforms, IRE1α and IRE1β, which both have RNase and kinase activities. However, in Arabidopsis thaliana, RNase activity of IRE1β, but not its protein kinase activity, is required for ER stress-mediated autophagy [73]. In Dictyostelium, the response to ER stress involves the combined activation of an IRE1α-dependent gene expression program and the autophagy pathway [74]. In mammalian cells, the spliced form of XBP 1 upregulates Nedd4-2, an E3 ubiquitin ligase involved in targeting proteins for subsequent degradation, in response to ER stress. It is also important for the induction of an appropriate autophagic response [75].

Different cancer cell models have allowed a better understanding of the mechanisms involved in autophagy triggered by ER stress. In cervical tumor cells, ER stress and UPR induced by X-ray exposition led to the activation of the NF-κB signaling pathway, autophagy, and apoptosis [76]. NF-κB is important for the proliferation, invasion, and metastasis of cervical cancer cells. Furthermore, in a model of breast cancer, autophagy and apoptosis were triggered through ER stress, UPR, and a high expression of CHOP and JNK [77].

Moreover, ERK and JNK activation is associated with cross talk between autophagy and another PCD. In L929 fibrosarcoma cells, ERK and JNK can link a signal from caspase-8 inhibition to autophagy, which in turn induce ROS production and PARP activation, leading to ATP depletion and necroptosis [78].

Ca2+ exchange between the ER and mitochondria is mediated through domains called mitochondria-associated membranes (MAMs). The interruption of Ca2+ flux between these organelles generates metabolic stress where AMPK present in MAMs triggers autophagy via beclin-1 phosphorylation [79, 80]. Autophagy activation might prevent proper interorganelle communication that would maintain mitochondrial function and cellular homeostasis [79].

In ER stress, some miRNAs promote the survival of the cells, while others promote cell death. In HeLa cells under RE stress, miR-346 positively regulates the expression of glycogen synthase kinase 3 beta (GSK3B) which reduces the interaction of beclin-1 and BCL2 to induce autophagy, ROS reduction, and cell death [81].

5.2 The role of the ER in autophagy during follicular atresia

Autophagy is mainly induced in granulosa cells (Figure 2c) during folliculogenesis and shows a high correlation with apoptosis, and furthermore, both routes of PCD could play active roles in oocyte depletion [82]. According to Meng et al. [83], antral follicular degeneration is initiated by granulosa cell apoptosis, while preantral follicular atresia occurs mainly via enhanced granulosa cell autophagy. Surprisingly, apoptosis and autophagy can be present in the same cell at the same time, just as cells can show caspase-3 active, DNA fragmentation, and immunodetection of LC3 and Lamp 1 [2, 3].

The signals that establish autophagy or apoptosis as the route of cell death are not fully understood. Consistent with Zhang et al. [84], atresia initiation is associated with a cross talk of different PCDs including apoptosis and autophagy, a dramatic shift of steroidogenic enzymes, deficient glutathione metabolism, and vascular degeneration. In a rat model, FSH, a survival factor, decreased autophagy through LC3-II inhibition and Akt-mTOR pathway activation [85]. Shen et al. [86] assessed the mechanism involved in autophagy inhibition by the Akt-mTOR pathway in granulosa cells exposed to FSH and oxidative stress because mTOR, a negative regulator of autophagy, inhibits FOXO1, which promotes the expression of several autophagy genes. They found that FSH induced granulosa cell survival via FOXO1 inhibition by the PI3K-Akt-mTOR pathway [86]. Nevertheless, in mouse granulosa cells, FSH was related to follicle development and atresia because FSH induces autophagy signaling via HIF-1α [87].

Despite the studies on the role of the ER in autophagy, its specific participation in follicular atresia is still unknown.

Advertisement

6. Necroptosis

Necroptosis is a subtype of regulated necrosis and shares the same morphological changes, including organelle swelling and membrane rupture. Necroptosis is a caspase-independent cell death, and its execution involves the active disintegration of mitochondrial, lysosomal, and plasma membranes. This PCD is triggered by various stimuli, such as TNF, Fas ligand, and TRAIL and depends on the serine/threonine kinase activity of RIP1. Additionally, a set of 432 genes regulates necroptosis and cellular sensitivity to this PCD by a signaling network that mediates innate immunity [88]. Moreover, Bmf, a BH3-only protein, is required for death receptor-induced necroptosis [88].

Moreover, environmental toxicants like cadmium can activate necroptosis. Intermediate levels of cadmium are associated with lost plasma membrane integrity, a decrease of ATP levels, and mitochondrial membrane potential and cell swelling, which are features associated with necroptotic cell death [89].

The core pathway of necroptosis relies on the assembly of an amyloid-like structure termed the necrosome. The necrosome is a multiprotein complex formed by receptor-interacting protein kinase 3 (RIPK3), RIPK1, and mixed lineage kinase domain-like (MLKL). Oligomerization and intramolecular autophosphorylation of RIPK3 lead to the recruitment and phosphorylation of MLKL. RIPK3 and MLKL continuously shuttle between the nucleus and the cytoplasm, whereas RIPK1 is constitutively present in both compartments [90]. Nuclear RIPK1 becomes ubiquitinated, and then nuclear MLKL becomes phosphorylated and oligomerized [90]. MLKL mediates plasma membrane rupture. MLKL forms cation channels that are preferentially permeable to Mg2+ in the presence of Na+ and K+ [91]. MLKL-induced membrane depolarization and cell death exhibit a positive correlation to channel activity.

6.1 The role of ER in necroptosis

The role of the ER in necroptosis has been evidenced using necrostatin-1, an inhibitor of necroptosis, which has a protective effect on the endoplasmic reticulum and mitochondria and alleviates ER stress after spinal cord injury [92]. Furthermore, Grp78 promotes an inflammatory response through the upregulation of necroptosis and subsequent activation of NF-κB and AP-1 pathways [93]. The depletion of reticulocalbin 1, an ER-resident Ca2+-binding protein, induces Grp78, activates PERK, and phosphorylates eIF2α. Moreover, the activation of CaMKII and the inactivation of Akt are important for necroptosis in response to reticulocalbin 1 depletion [94].

The function of MLKL and RIPK in necroptosis has been widely studied. The signal transducer and activator of transcription 3 (STAT3) was demonstrated to be downstream of calpain and regulates RIPK3 expression and MLKL phosphorylation and induces ER stress and mitochondrial calcium dysregulation [95]. Moreover, in cardiomyocytes upregulated RIPK1 and RIPK3 evoke ER stress, accompanied by an increase in intracellular Ca2+ levels and xanthine oxidase expression, which raised cellular ROS that mediated the mitochondrial permeability transition pore opening and necroptosis [96, 97]. In addition, the activation of JNK1/2 is regulated by RIPK3 [96].

Moreover, there are proteins that can participate in necroptosis and other types of PCD such as AIF and MLKL. Apoptosis-inducing factor (AIF), a protein normally located within the intermembrane space of mitochondria, is linked to apoptosis and necrosis. However, it has been shown that mitochondrial depolarization induced by ER stress promotes AIF release and nuclear condensation, which is consistent with necroptotic cell death [98, 99, 100]. MLKL, a member of the necrosome, also participates in chelerythrine (CHE)-promoted apoptosis through nuclear MLKL translocation and a special band of MLKL, which is promoted by a mutual regulation between the MLKL and PERK-eIF2α pathways in response to ROS formation [101].

6.2 The role of the RE in necroptosis during follicular atresia

Necroptosis has been widely researched, but there is still much to investigate, including the mechanism that mediates its execution. Nevertheless, necroptosis studies have been carried out under pathological conditions, and thus it is important to use physiological models like follicular atresia.

Necroptosis contributes to follicular atresia and luteolysis [102]. The factors involved in granulosa cell necroptosis can be regulated by acetylcholinesterase (AChE), cytokines, starvation, and oxidative stress via TNFα [103]. Also, an ovarian AChE variant, the read-through isoform AChE-R, has a nonenzymatic function that stimulates RIPK1-/MLKL-dependent necroptosis [103]. Therefore, although the participation of the ER in necroptosis and the contribution of this PCD in follicular atresia have been shown, the interrelation between ER stress-induced necroptosis and follicular atresia is completely unknown.

Advertisement

7. Paraptosis

Sperandio et al. [104] introduced the term paraptosis to describe a route of caspase-independent PCD that has morphological, biochemical, and transcriptional features that are different from apoptosis [104]. Endoplasmic reticulum swelling, mitochondrial swelling, and resistance to apoptosis inhibitors without nuclear shrinkage or pyknosis characterize paraptosis. Although paraptosis is a caspase-independent cell death, participation of caspase-9 has been shown under experimental conditions [104].

Paraptosis can be triggered by different stimuli including insulin-like growth factor I receptor (IGFIR), JAY/TROY, and ROS. IGF-I is a regulator of multiple cell signaling pathways including PI3K-Akt1-RPS6 and ERK1/2 MAPK that are critical for cell proliferation, migration, and survival [105]. IGFIR-induced paraptosis is mediated by caspase-9, and at least two signal transduction pathways participate in the execution of paraptosis, the MAPK and JNK pathways [104, 106].

TAJ/TROY, a member of the tumor necrosis factor receptor superfamily, induces morphological features of paraptosis accompanied by phosphatidylserine externalization, the loss of the mitochondrial transmembrane potential, and independent caspase activation [105]. Moreover, programmed cell death 5 (PDCD5), an apoptosis-promoting protein, enhances TAJ-/TROY-induced paraptotic cell death [107].

ROS production can trigger paraptosis through PINK and mitophagy activation [108, 109]. Covalent modifications of free sulfhydryl groups on proteins cause protein misfolding and the accumulation of misfolded proteins, leading to ER stress, CHOP activation, and paraptosis [110, 111]. In malignant hepatoma cells with Bcl-xL-mediated apoptotic defects, the disruption of thiol homeostasis and treatment with doxorubicin and pyrrolidine dithiocarbamate induced paraptotic cell death [112].

The full signal transduction pathway and identification of specific markers for paraptosis are still unclear. Nevertheless, phosphatidylethanolamine-binding protein (PEBP-1), a suppressor of the MAPK pathway, has been identified, and prohibitin, a mitochondrial protein, is a mediator of paraptosis [113]. Furthermore, the redistribution of α- and β-tubulin and tropomyosin has been observed in the early stages of paraptosis. Other characteristics of the paraptotic pathway involve alterations mainly in signal transduction proteins, mitochondrial proteins, and some metabolic proteins [113].

7.1 The role of RE in paraptosis

Cancer cells are the best model to study paraptosis because there can be apoptosis and/or autophagy resistance. In melanoma cells, the sustained activation of the IRE1α and ATF6 pathways driven by the MEK/ERK pathway avoids ER stress-induced apoptosis [114].

Different compounds for cancer treatment have shown paraptosis induction. For example, HeLa, A549, and PC-3 cells treated with celastrol induced vacuoles derived from the dilation of ER, a feature of apoptotic cell death; moreover, this was accompanied by autophagy and apoptosis. Furthermore, the ER swelling triggered by celastrol induced ER stress markers including Grp78, PERK, IRE, and CHOP and alterations to proteasome function that resulted in the accumulation of ubiquitinated protein [115, 116]. Moreover, paraptosis can be accelerated by pre-treatment with the proteasome inhibitor MG132 [117]. On the other hand, cyclosporine A treatment of cervical cancerous SiHa cells showed ER stress and UPR preceded by massive cytoplasmic vacuole formation that culminated in a paraptosis-like cell death [118]. Moreover, murine hepatoma 1c1c7 cells and the human non-small cell lung cancer A549 cell line exposed to a combination of photodamage and benzoporphyrin derivative result in ER swelling and paraptotic cell death [119].

For the pathways involved in paraptosis, ER vacuoles can be dependent on the PI3K/Akt signaling pathway [120]. Moreover, in BC3H1 myoblast cell lines exposed to yessotoxin, paraptosis was accompanied by cytoskeletal alterations and the activation of JNK/SAPK1 [121]. However, in acute lymphoblastic leukemia cells, everolimus, a mTOR inhibitor, showed that JNK signaling was not required for paraptotic cell death [122]. Paraptosis in epithelial ovarian cancer (EOC) cells treated with morusin was characterized by VDAC-mediated Ca2+ influx into mitochondria, and subsequent mitochondrial Ca2+ overload contributes to mitochondrial swelling and dysfunction, leading to the accumulation of ER stress markers, the generation of ROS, and the loss of mitochondrial membrane potential (Δψm) in EOC cells [123].

7.2 The role of the RE in paraptosis during follicular atresia

Knowledge of the role of paraptosis during follicular atresia is still limited. In Bombyx mori, apoptosis, autophagy, and paraptosis occur in the ovarian nurse cell cluster during late vitellogenesis, whereas middle vitellogenesis is exclusively characterized by the presence of paraptosis, preceding both apoptosis and autophagy [124]. In mammals, paraptosis was evidenced by ER swelling (Figure 2d) and CHOP immunodetection in granulosa cells during follicular atresia in adult Wistar rats [4].

The mechanisms involved in paraptosis during follicular atresia are still unknown. The paraptotic inductor IGFR might be related because it is implicated in follicular growth and selection [104, 125]. Moreover, IGF2R and the binding protein genes IGFBP5 and IGFBP6 are overexpressed in atretic follicles [126]. However, more studies on paraptosis during follicular atresia are necessary.

Advertisement

8. Conclusions

Endoplasmic reticulum stress is a strong signal that triggers different programmed cell death pathways. Interestingly, programmed cell death via endoplasmic reticulum stress is not exclusive to pathological or experimental conditions but is present in physiological processes like follicular atresia. However, the specific mechanisms and signals for choosing a particular cell death pathway are still unknown. In this way, research on the pathways and mechanisms involved in programmed cell death activated by endoplasmic reticulum stress are fundamental, particularly for follicular atresia, as this process ensures the ovulation of competent oocytes for fertilization.

Advertisement

Acknowledgments

This work was supported by PAPIIT IN225117 and PAPIIT IN227919.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Hsu CJ, Hammond JM. Gonadotropins and estradiol stimulate immunoreactive insulin-like growth factor-I production by porcine granulosa cells in vitro. Endocrinology. 1987;120(1):198-207. DOI: 10.1210/endo-120-1-198
  2. 2. Escobar Sánchez ML, Echeverría Martínez OM, Vázquez-Nin GH. Immunohistochemical and ultrastructural visualization of different routes of oocyte elimination in adult rats. European Journal of Histochemistry. 2012;56(2):e17. DOI: 10.4081/ejh.2012.17
  3. 3. Escobar ML, Echeverría OM, Casasa AS, García G, Aguilar SJ, Vázquez-Nin GH. Involvement of pro-apoptotic and pro-autophagic proteins in granulosa cell death. Cell Biology. 2013;1(1):9-17. DOI: 10.11648/j.cb.20130101.12
  4. 4. Torres-Ramírez N, Escobar ML, Vázquez-Nin GH, Ortiz R, Echeverría OM. Paraptosis-like cell death in Wistar rat granulosa cells. Development, Growth & Differentiation. 2016;58(8):651-663. DOI: 10.1111/dgd.12322
  5. 5. Brodsky JL. Cleaning up: ER-associated degradation to the rescue. Cell. 2012;151(6):1163-1167. DOI: 10.1016/j.cell.2012.11.012
  6. 6. Cox JS, Shamu CE, Walter P. Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase. Cell. 1993;73(6):1197-1206
  7. 7. Walter P, Ron D. The unfolded protein response: From stress pathway to homeostatic regulation. Science. 2011;334(6059):1081-1086. DOI: 10.1126/science.1209038
  8. 8. Karagöz GE, Acosta-Alvear D, Nguyen HT, Lee CP, Chu F, Walter P. An unfolded protein-induced conformational switch activates mammalian IRE1. eLife. 2017;6:pii: e30700. DOI: 10.7554/eLife.30700
  9. 9. Su Q, Wang S, Gao HQ, Kazemi S, Harding HP, Ron D, et al. Modulation of the eukaryotic initiation factor 2 alpha-subunit kinase PERK by tyrosine phosphorylation. The Journal of Biological Chemistry. 2008;283(1):469-475. DOI: 10.1074/jbc.M704612200
  10. 10. Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, et al. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002;415(6867):92-96. DOI: 10.1038/415092a
  11. 11. Hollien J, Lin JH, Li H, Stevens N, Walter P, Weissman JS. Regulated Ire1-dependent decay of messenger RNAs in mammalian cells. The Journal of Cell Biology. 2009;186(3):323-331. DOI: 10.1083/jcb.200903014
  12. 12. Lee AH, Iwakoshi NN, Glimcher LH. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Molecular and Cellular Biology. 2003;23(21):7448-7459
  13. 13. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nature Cell Biology. 2000;2(6):326-332. DOI: 10.1038/35014014
  14. 14. Harding HP, Zhang Y, Bertolotti A, Zeng H, Ron D. Perk is essential for translational regulation and cell survival during the unfolded protein response. Molecular Cell. 2000;5(5):897-904
  15. 15. Luo S, Mao C, Lee B, Lee AS. GRP78/BiP is required for cell proliferation and protecting the inner cell mass from apoptosis during early mouse embryonic development. Molecular and Cellular Biology. 2006;26(15):5688-5697. DOI: 10.1128/MCB.00779-06
  16. 16. Schindler AJ, Schekman R. In vitro reconstitution of ER-stress induced ATF6 transport in COPII vesicles. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(42):17775-17780. DOI: 10.1073/pnas.0910342106
  17. 17. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 2001;107(7):881-891
  18. 18. Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, et al. IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge regulate XBP1 in signalling the unfolded protein response. Genes & Development. 2002;16(4):452-466. DOI: 10.1101/gad.964702
  19. 19. Lombardi A, Ulianich L, Treglia AS, Nigro C, Parrillo L, Lofrumento DD, et al. Increased hexosamine biosynthetic pathway flux dedifferentiates INS-1E cells and murine islets by an extracellular signal-regulated kinase (ERK)1/2-mediated signal transmission pathway. Diabetologia. 2012;55(1):141-153. DOI: 10.1007/s00125-011-2315-1
  20. 20. Henderson KM, McNatty KP, Smith P, Gibb M, O'Keeffe LE, Lun S, et al. Influence of follicular health on the steroidogenic and morphological characteristics of bovine granulosa cells in vitro. Journal of Reproduction and Fertility. 1987;79(1):185-193
  21. 21. Kovács J, Forgó V, Péczely P. The fine structure of the follicular cells in growing and atretic ovarian follicles of the domestic goose. Cell and Tissue Research. 1992;267(3):561-569
  22. 22. Huang N, Yu Y, Qiao J. Dual role for the unfolded protein response in the ovary: Adaption and apoptosis. Protein & Cell. 2017;8(1):14-24. DOI: 10.1007/s13238-016-0312-3
  23. 23. Harada M, Nose E, Takahashi N, Hirota Y, Hirata T, Yoshino O, et al. Evidence of the activation of unfolded protein response in granulosa and cumulus cells during follicular growth and maturation. Gynecological Endocrinology. 2015;31(10):783-787. DOI: 10.3109/09513590.2015.1062862
  24. 24. Yang X, Wu LL, Chura LR, Liang X, Lane M, Norman RJ, et al. Exposure to lipid-rich follicular fluid is associated with endoplasmic reticulum stress and impaired oocyte maturation in cumulus-oocyte complexes. Fertility and Sterility. 2012;97(6):1438-1443. DOI: 10.1016/j.fertnstert.2012.02.034
  25. 25. Lumley EC, Osborn AR, Scott JE, Scholl AG, Mercado V, McMahan YT, et al. Moderate endoplasmic reticulum stress activates a PERK and p38-dependent apoptosis. Cell Stress & Chaperones. 2017;22(1):43-54. DOI: 10.1007/s12192-016-0740-2
  26. 26. Kerr JF, Wyllie AH, Currie AR. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. British Journal of Cancer. 1972;26(4):239-257
  27. 27. Fraser A, Evan G. A license to kill. Cell. 1996;85(6):781-784
  28. 28. Hengartner MO. The biochemistry of apoptosis. Nature. 2000;407(6805):770-776. DOI: 10.1038/35037710
  29. 29. Strasser A, O'Connor L, Dixit VM. Apoptosis signaling. Annual Review of Biochemistry. 2000;69:217-245. DOI: 10.1146/annurev.biochem.69.1.217
  30. 30. Jeng PS, Inoue-Yamauchi A, Hsieh JJ, Cheng EH. BH3-dependent and independent activation of BAX and BAK in mitochondrial apoptosis. Current Opinion in Physiology. 2018;3:71-81. DOI: 10.1016/j.cophys.2018.03.005
  31. 31. Holinger EP, Chittenden T, Lutz RJ. Bak BH3 peptides antagonize Bcl-xL function and induce apoptosis through cytochrome c-independent activation of caspases. The Journal of Biological Chemistry. 1999;274(19):13298-13304
  32. 32. Narita M, Shimizu S, Ito T, Chittenden T, Lutz RJ, Matsuda H, et al. Bax interacts with the permeability transition pore to induce permeability transition and cytochrome c release in isolated mitochondria. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(25):14681-14686
  33. 33. Bröker LE, Kruyt FA, Giaccone G. Cell death independent of caspases: A review. Clinical Cancer Research. 2005;11(9):3155-3162. DOI: 10.1158/1078-0432.CCR-04-2223
  34. 34. Tilly JL, Kowalski KI, Johnson AL, Hsueh AJ. Involvement of apoptosis in ovarian follicular atresia and postovulatory regression. Endocrinology. 1991;129(5):2799-2801. DOI: 10.1210/endo-129-5-2799
  35. 35. Hussein MR. Apoptosis in the ovary: Molecular mechanisms. Human Reproduction Update. 2005;11(2):162-177. DOI: 10.1093/humupd/dmi001
  36. 36. Coucouvanis EC, Sherwood SW, Carswell-Crumpton C, Spack EG, Jones PP. Evidence that the mechanism of prenatal germ cell death in the mouse is apoptosis. Experimental Cell Research. 1993;209(2):238-247. DOI: 10.1006/excr.1993.1307
  37. 37. Escobar ML, Echeverría OM, Sánchez-Sánchez L, Méndez C, Pedernera E, Vázquez-Nin GH. Analysis of different cell death processes of prepubertal rat oocytes in vitro. Apoptosis. 2010;15(4):511-526. DOI: 10.1007/s10495-009-0448-1
  38. 38. Poljicanin A, Vukusic Pusic T, Vukojevic K, Caric A, Vilovic K, Tomic S, et al. The expression patterns of pro-apoptotic and anti-apoptotic factors in human fetal and adult ovary. Acta Histochemica. 2013;115(6):533-540. DOI: 10.1016/j.acthis.2012.11.009
  39. 39. Darling NJ, Balmanno K, Cook SJ. ERK1/2 signalling protects against apoptosis following endoplasmic reticulum stress but cannot provide long-term protection against BAX/BAK-independent cell death. PLoS One. 2017;12(9):e0184907. DOI: 10.1371/journal.pone.0184907
  40. 40. Urra H, Dufey E, Lisbona F, Rojas-Rivera D, Hetz C. When ER stress reaches a dead end. Biochimica et Biophysica Acta. 2013;1833(12):3507-3517. DOI: 10.1016/j.bbamcr.2013.07.024
  41. 41. Hetz C, Bernasconi P, Fisher J, Lee AH, Bassik MC, Antonsson B, et al. Proapoptotic BAX and BAK modulate the unfolded protein response by a direct interaction with IRE1alpha. Science. 2006;312(5773):572-576. DOI: 10.1126/science.1123480
  42. 42. Gupta S, Read DE, Deepti A, Cawley K, Gupta A, Oommen D, et al. Perk-dependent repression of miR-106b-25 cluster is required for ER stress-induced apoptosis. Cell Death & Disease. 2012;3:e333. DOI: 10.1038/cddis.2012.74
  43. 43. Puthalakath H, O'Reilly LA, Gunn P, Lee L, Kelly PN, Huntington ND, et al. ER stress triggers apoptosis by activating BH3-only protein Bim. Cell. 2007;129(7):1337-1349. DOI: 10.1016/j.cell.2007.04.027
  44. 44. Li J, Lee B, Lee AS. Endoplasmic reticulum stress-induced apoptosis: Multiple pathways and activation of p53-up-regulated modulator of apoptosis (PUMA) and NOXA by p53. The Journal of Biological Chemistry. 2006;281(11):7260-7270. DOI: 10.1074/jbc.M509868200
  45. 45. Sánchez AM, Martínez-Botas J, Malagarie-Cazenave S, Olea N, Vara D, Lasunción MA, et al. Induction of the endoplasmic reticulum stress protein GADD153/CHOP by capsaicin in prostate PC-3 cells: A microarray study. Biochemical and Biophysical Research Communications. 2008;372(4):785-791. DOI: 10.1016/j.bbrc.2008.05.138
  46. 46. Han J, Back SH, Hur J, Lin YH, Gildersleeve R, Shan J, et al. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nature Cell Biology. 2013;15(5):481-490. DOI: 10.1038/ncb2738
  47. 47. Upton JP, Wang L, Han D, Wang ES, Huskey NE, Lim L, et al. IRE1α cleaves select microRNAs during ER stress to derepress translation of proapoptotic caspase-2. Science. 2012;338(6108):818-822. DOI: 10.1126/science.1226191
  48. 48. Wu Y, Ma C, Zhao H, Zhou Y, Chen Z, Wang L. Alleviation of endoplasmic reticulum stress protects against cisplatin-induced ovarian damage. Reproductive Biology and Endocrinology. 2018;16(1):85. DOI: 10.1186/s12958-018-0404-4
  49. 49. Lin P, Yang Y, Li X, Chen F, Cui C, Hu L, et al. Endoplasmic reticulum stress is involved in granulosa cell apoptosis during follicular atresia in goat ovaries. Molecular Reproduction and Development. 2012;79(6):423-432. DOI: 10.1002/mrd.22045
  50. 50. Yang Y, Lin P, Chen F, Wang A, Lan X, Song Y, et al. Luman recruiting factor regulates endoplasmic reticulum stress in mouse ovarian granulosa cell apoptosis. Theriogenology. 2013;79(4):633-639. DOI: 10.1016/j.theriogenology.2012.11.017
  51. 51. Xiong Y, Chen H, Lin P, Wang A, Wang L, Jin Y. ATF6 knockdown decreases apoptosis, arrests the S phase of the cell cycle, and increases steroid hormone production in mouse granulosa cells. American Journal of Physiology. Cell Physiology. 2017;312(3):C341-C353. DOI: 10.1152/ajpcell.00222.2016
  52. 52. Yang D, Jiang T, Lin P, Chen H, Wang L, Wang N, et al. Knock-down of apoptosis inducing factor gene protects endoplasmic reticulum stress-mediated goat granulosa cell apoptosis. Theriogenology. 2017;88:89-97. DOI: 10.1016/j.theriogenology.2016.10.001
  53. 53. Santos CX, Tanaka LY, Wosniak J, Laurindo FR. Mechanisms and implications of reactive oxygen species generation during the unfolded protein response: Roles of endoplasmic reticulum oxidoreductases, mitochondrial electron transport, and NADPH oxidase. Antioxidants & Redox Signaling. 2009;11(10):2409-2427. DOI: 10.1089/ars.2009.2625
  54. 54. Liu ZQ , Shen M, Wu WJ, Li BJ, Weng QN, Li M, et al. Expression of PUMA in follicular granulosa cells regulated by FoxO1 activation during oxidative stress. Reproductive Sciences. 2015;22(6):696-705. DOI: 10.1177/1933719114556483
  55. 55. Matsumine M, Shibata N, Ishitani K, Kobayashi M, Ohta H. Pentosidine accumulation in human oocytes and their correlation to age-related apoptosis. Acta Histochemica et Cytochemica. 2008;41(4):97-104. DOI: 10.1267/ahc.08014
  56. 56. Park HJ, Park SJ, Koo DB, Lee SR, Kong IK, Ryoo JW, et al. Progesterone production is affected by unfolded protein response (UPR) signalling during the luteal phase in mice. Life Sciences. 2014;113(1-2):60-67. DOI: 10.1016/j.lfs.2014.07.033
  57. 57. Yang Y, Sun M, Shan Y, Zheng X, Ma H, Ma W, et al. Endoplasmic reticulum stress-mediated apoptotic pathway is involved in corpus luteum regression in rats. Reproductive Sciences. 2015;22(5):572-584. DOI: 10.1177/1933719114553445
  58. 58. Kunz JB, Schwarz H, Mayer A. Determination of four sequential stages during microautophagy in vitro. The Journal of Biological Chemistry. 2004;279(11):9987-9996. DOI: 10.1074/jbc.M307905200
  59. 59. Chiang HL, Terlecky SR, Plant CP, Dice JF. A role for a 70-kilodalton heat shock protein in lysosomal degradation of intracellular proteins. Science. 1989;246(4928):382-385
  60. 60. Cuervo AM, Dice JF. A receptor for the selective uptake and degradation of proteins by lysosomes. Science. 1996;273(5274):501-503
  61. 61. Kaushik S, Kiffin R, Cuervo AM. Chaperone-mediated autophagy and aging: A novel regulatory role of lipids revealed. Autophagy. 2007;3(4):387-389
  62. 62. Dunn WA Jr. Autophagy and related mechanisms of lysosome-mediated protein degradation. Trends in Cell Biology. 1994;4(4):139-143
  63. 63. Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins in autophagosome formation. Annual Review of Cell and Developmental Biology. 2011;27:107-132. DOI: 10.1146/annurev-cellbio-092910-154005
  64. 64. Olsvik HL, Lamark T, Takagi K, Larsen KB, Evjen G, Øvervatn A, et al. FYCO1 contains a C-terminally extended, LC3A/B-preferring LC3-interacting region (LIR) motif required for efficient maturation of autophagosomes during basal autophagy. The Journal of Biological Chemistry. 2015;290(49):29361-29374. DOI: 10.1074/jbc.M115.686915
  65. 65. Pankiv S, Alemu EA, Brech A, Bruun JA, Lamark T, Overvatn A, et al. FYCO1 is a Rab7 effector that binds to LC3 and PI3P to mediate microtubule plus end-directed vesicle transport. The Journal of Cell Biology. 2010;188(2):253-269. DOI: 10.1083/jcb.200907015
  66. 66. Yang D, Jiang T, Liu J, Hong J, Lin P, Chen H, et al. Hormone regulates endometrial function via cooperation of endoplasmic reticulum stress and mTOR-autophagy. Journal of Cellular Physiology. 2018;233(9):6644-6659. DOI: 10.1002/jcp.26315
  67. 67. Ishida Y, Yamamoto A, Kitamura A, Lamandé SR, Yoshimori T, Bateman JF, et al. Autophagic elimination of misfolded procollagen aggregates in the endoplasmic reticulum as a means of cell protection. Molecular Biology of the Cell. 2009;20(11):2744-2754. DOI: 10.1091/mbc.e08-11-1092
  68. 68. Ren B, Wang Y, Wang H, Wu Y, Li J, Tian J. Comparative proteomics reveals the neurotoxicity mechanism of ER stressors tunicamycin and dithiothreitol. Neurotoxicology. 2018;68:25-37. DOI: 10.1016/j.neuro.2018.07.004
  69. 69. Yorimitsu T, Nair U, Yang Z, Klionsky DJ. Endoplasmic reticulum stress triggers autophagy. The Journal of Biological Chemistry. 2006;281(40):30299-30304. DOI: 10.1074/jbc.M607007200
  70. 70. Fumagalli F, Noack J, Bergmann TJ, Cebollero E, Pisoni GB, Fasana E, et al. Translocon component Sec62 acts in endoplasmic reticulum turnover during stress recovery. Nature Cell Biology. 2016;18(11):1173-1184. DOI: 10.1038/ncb3423
  71. 71. B'chir W, Maurin AC, Carraro V, Averous J, Jousse C, Muranishi Y, et al. The eIF2α/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Research. 2013;41(16):7683-7699. DOI: 10.1093/nar/gkt563
  72. 72. Jensen BC, Bultman SJ, Holley D, Tang W, de Ridder G, Pizzo S, et al. Upregulation of autophagy genes and the unfolded protein response in human heart failure. International Journal of Clinical and Experimental Medicine. 2017;10(1):1051-1058
  73. 73. Bao Y, Pu Y, Yu X, Gregory BD, Srivastava R, Howell SH, et al. IRE1B degrades RNAs encoding proteins that interfere with the induction of autophagy by ER stress in Arabidopsis thaliana. The FASEB Journal. 2016;30(9):3053-3068. DOI: 10.1080/15548627.2018.1462426
  74. 74. Domínguez-Martín E, Ongay-Larios L, Kawasaki L, Vincent O, Coello G, Coria R, et al. IreA controls endoplasmic reticulum stress-induced autophagy and survival through homeostasis recovery. Molecular and Cellular Biology. 2018;pii: MCB.00054-pii: MCB.00018. DOI: 10.1128/MCB.00054-18
  75. 75. Wang H, Sun RQ , Camera D, Zeng XY, Jo E, Chan SM, et al. Endoplasmic reticulum stress up-regulates Nedd4-2 to induce autophagy. The FASEB Journal. 2016;30(7):2549-2556. DOI: 10.1096/fj.201500119
  76. 76. Zhu X, Huang L, Gong J, Shi C, Wang Z, Ye B, et al. NF-κB pathway link with ER stress-induced autophagy and apoptosis in cervical tumor cells. Cell Death Discovery. 2017;3:17059. DOI: 10.1038/cddiscovery.2017.59
  77. 77. Li F, Zheng X, Liu Y, Li P, Liu X, Ye F, et al. Different roles of CHOP and JNK in mediating radiation-induced autophagy and apoptosis in breast cancer cells. Radiation Research. 2016;185(5):539-548. DOI: 10.1667/RR14344.1
  78. 78. Chen SY, Chiu LY, Maa MC, Wang JS, Chien CL, Lin WW. zVAD-induced autophagic cell death requires c-Src-dependent ERK and JNK activation and reactive oxygen species generation. Autophagy. 2011;7(2):217-228
  79. 79. Ahumada-Castro U, Silva-Pavez E, Lovy A, Pardo E, Molgo J, Cárdenas C. MTOR-independent autophagy induced by interrupted endoplasmic reticulum-mitochondrial Ca2+ communication: A dead end in cancer cells. Autophagy. 2018. DOI: 10.1080/15548627.2018.1537769
  80. 80. Zhang D, Wang W, Sun X, Xu D, Wang C, Zhang Q , et al. AMPK regulates autophagy by phosphorylating BECN1 at threonine 388. Autophagy. 2016;12(9):1447-1459. DOI: 10.1080/15548627.2016.1185576
  81. 81. Guo J, Yang Z, Yang X, Li T, Liu M, Tang H. miR-346 functions as a pro-survival factor under ER stress by activating mitophagy. Cancer Letters. 2018;413:69-81. DOI: 10.1016/j.canlet.2017.10.030
  82. 82. Yadav PK, Tiwari M, Gupta A, Sharma A, Prasad S, Pandey AN, et al. Germ cell depletion from mammalian ovary: Possible involvement of apoptosis and autophagy. Journal of Biomedical Science. 2018;25(1):36. DOI: 10.1186/s12929-018-0438-0
  83. 83. Meng L, Jan SZ, Hamer G, van Pelt AM, van der Stelt I, Keijer J, et al. Preantral follicular atresia occurs mainly through autophagy, while antral follicles degenerate mostly through apoptosis. Biology of Reproduction. 2018;99(4):853-863. DOI: 10.1093/biolre/ioy116
  84. 84. Zhang J, Liu Y, Yao W, Li Q , Liu H, Pan Z. Initiation of follicular atresia: Gene networks during early atresia in pig ovaries. Reproduction. 2018;156(1):23-33. DOI: 10.1530/REP-18-0058
  85. 85. Choi J, Jo M, Lee E, Choi D. AKT is involved in granulosa cell autophagy regulation via mTOR signaling during rat follicular development and atresia. Reproduction. 2013;147(1):73-80. DOI: 10.1530/REP-13-0386
  86. 86. Shen M, Jiang Y, Guan Z, Cao Y, Li L, Liu H, et al. Protective mechanism of FSH against oxidative damage in mouse ovarian granulosa cells by repressing autophagy. Autophagy. 2017;13(8):1364-1385. DOI: 10.1080/15548627.2017.1327941
  87. 87. Zhou J, Yao W, Li C, Wu W, Li Q , Liu H. Administration of follicle-stimulating hormone induces autophagy via upregulation of HIF-1α in mouse granulosa cells. Cell Death & Disease. 2017;8(8):e3001. DOI: 10.1038/cddis.2017.371
  88. 88. Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, Xavier RJ, et al. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell. 2008;135(7):1311-1323. DOI: 10.1016/j.cell.2008.10.044
  89. 89. Krumschnabel G, Ebner HL, Hess MW, Villunger A. Apoptosis and necroptosis are induced in rainbow trout cell lines exposed to cadmium. Aquatic Toxicology. 2010;99(1):73-85. DOI: 10.1016/j.aquatox.2010.04.005
  90. 90. Weber K, Roelandt R, Bruggeman I, Estornes Y, Vandenabeele P. Nuclear RIPK3 and MLKL contribute to cytosolic necrosome formation and necroptosis. Communications Biology. 2018;1:6. DOI: 10.1038/s42003-017-0007-1
  91. 91. Xia B, Fang S, Chen X, Hu H, Chen P, Wang H, et al. MLKL forms cation channels. Cell Research. 2016;26(5):517-528. DOI: 10.1038/cr.2016.26
  92. 92. Wang S, Wu J, Zeng YZ, Wu SS, Deng GR, Chen ZD, et al. Necrostatin-1 mitigates endoplasmic reticulum stress after spinal cord injury. Neurochemical Research. 2017;42(12):3548-3558. DOI: 10.1007/s11064-017-2402-x
  93. 93. Wang Y, Zhou JS, Xu XC, Li ZY, Chen HP, Ying SM, et al. Endoplasmic reticulum chaperone GRP78 mediates cigarette smoke-induced necroptosis and injury in bronchial epithelium. International Journal of Chronic Obstructive Pulmonary Disease. 2018;13:571-581. DOI: 10.2147/COPD.S150633
  94. 94. Liu X, Zhang N, Wang D, Zhu D, Yuan Q , Zhang X, et al. Downregulation of reticulocalbin-1 differentially facilitates apoptosis and necroptosis in human prostate cancer cells. Cancer Science. 2018;109(4):1147-1157. DOI: 10.1111/cas.13541
  95. 95. Kim H, Zamel R, Bai XH, Lu C, Keshavjee S, Keshavjee S, et al. Ischemia-reperfusion induces death receptor-independent necroptosis via calpain-STAT3 activation in a lung transplant setting. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2018;315(4):L595-L608. DOI: 10.1152/ajplung.00069.2018
  96. 96. Jia Y, Wang F, Guo Q , Li M, Wang L, Zhang Z, et al. Curcumol induces RIPK1/RIPK3 complex-dependent necroptosis via JNK1/2-ROS signaling in hepatic stellate cells. Redox Biology. 2018;19:375-387. DOI: 10.1016/j.redox.2018.09.007
  97. 97. Zhu P, Hu S, Jin Q , Li D, Tian F, Toan S, et al. Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biology. 2018;16:157-168. DOI: 10.1016/j.redox.2018.02.019
  98. 98. Bastian A, Thorpe JE, Disch BC, Bailey-Downs LC, Gangjee A, Devambatla RK, et al. A small molecule with anticancer and antimetastatic activities induces rapid mitochondrial-associated necrosis in breast cancer. The Journal of Pharmacology and Experimental Therapeutics. 2015;353(2):392-404. DOI: 10.1124/jpet.114.220335
  99. 99. Leon LJ, Pasupuleti N, Gorin F, Carraway KL 3rd. A cell-permeant amiloride derivative induces caspase-independent, AIF-mediated programmed necrotic death of breast cancer cells. PLoS One. 2013;8(4):e63038. DOI: 10.1371/journal.pone.0063038
  100. 100. Pasupuleti N, Leon L, Carraway KL 3rd, Gorin F. 5-Benzylglycinyl-amiloride kills proliferating and nonproliferating malignant glioma cells through caspase-independent necroptosis mediated by apoptosis-inducing factor. The Journal of Pharmacology and Experimental Therapeutics. 2013;344(3):600-615. DOI: 10.1124/jpet.112.200519
  101. 101. Cao WX, Li T, Tang ZH, Zhang LL, Wang ZY, Guo X, et al. MLKL mediates apoptosis via a mutual regulation with PERK/eIF2α pathway in response to reactive oxygen species generation. Apoptosis. 2018;23(9-10):521-531. DOI: 10.1007/s10495-018-1475-6
  102. 102. Blohberger J, Kunz L, Einwang D, Berg U, Berg D, Ojeda SR, et al. Readthrough acetylcholinesterase (AChE-R) and regulated necrosis: Pharmacological targets for the regulation of ovarian functions? Cell Death & Disease. 2015;6:e1685. DOI: 10.1038/cddis.2015.51
  103. 103. Chaudhary GR, Yadav PK, Yadav AK, Tiwari M, Gupta A, Sharma A, et al. Necrosis and necroptosis in germ cell depletion from mammalian ovary. Journal of Cellular Physiology. 2018. DOI: 10.1002/jcp.27562
  104. 104. Sperandio S, de Belle I, Bredesen DE. An alternative, nonapoptotic form of programmed cell death. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(26):14376-14381. DOI: 10.1073/pnas.97.26.14376
  105. 105. Jeong W, Song G, Bazer FW, Kim J. Insulin-like growth factor I induces proliferation and migration of porcine trophectoderm cells through multiple cell signaling pathways, including protooncogenic protein kinase 1 and mitogen-activated protein kinase. Molecular and Cellular Endocrinology. 2014;384(1-2):175-184. DOI: 10.1016/j.mce.2014.01.023
  106. 106. Sperandio S, Poksay K, de Belle I, Lafuente MJ, Liu B, Nasir J, et al. Paraptosis: Mediation by MAP kinases and inhibition by AIP-1/Alix. Cell Death and Differentiation. 2004;11(10):1066-1075. DOI: 10.1038/sj.cdd.4401465
  107. 107. Wang Y, Li X, Wang L, Ding P, Zhang Y, Han W, et al. An alternative form of paraptosis-like cell death triggered by TAJ/TROY and enhanced by PDCD5 overexpression. Journal of Cell Science. 2004;117(Pt 8):1525-1532. DOI: 10.1242/jcs.00994
  108. 108. Han H, Chou CC, Li R, Liu J, Zhang L, Zhu W, et al. Chalcomoracin is a potent anticancer agent acting through triggering oxidative stress via a mitophagy- and paraptosis-dependent mechanism. Scientific Reports. 2018;8(1):9566. DOI: 10.1038/s41598-018-27724-3
  109. 109. Petrillo S, Chiabrando D, Genova T, Fiorito V, Ingoglia G, Vinchi F, et al. Heme accumulation in endothelial cells impairs angiogenesis by triggering paraptosis. Cell Death and Differentiation. 2018;25(3):573-588. DOI: 10.1038/s41418-017-0001-7
  110. 110. Hager S, Korbula K, Bielec B, Grusch M, Pirker C, Schosserer M, et al. The thiosemicarbazone Me2NNMe2 induces paraptosis by disrupting the ER thiol redox homeostasis based on protein disulfide isomerase inhibition. Cell Death & Disease. 2018;9(11):1052. DOI: 10.1038/s41419-018-1102-z
  111. 111. Kim IY, Kwon M, Choi MK, Lee D, Lee DM, Seo MJ, et al. Ophiobolin a kills human glioblastoma cells by inducing endoplasmic reticulum stress via disruption of thiol proteostasis. Oncotarget. 2017;8(63):106740-106752. DOI: 10.18632/oncotarget.22537
  112. 112. Park SS, Lee DM, Lim JH, Lee D, Park SJ, Kim HM, et al. Pyrrolidine dithiocarbamate reverses Bcl-xL-mediated apoptotic resistance to doxorubicin by inducing paraptosis. Carcinogenesis. 2018;39(3):458-470. DOI: 10.1093/carcin/bgy003
  113. 113. Sperandio S, Poksay KS, Schilling B, Crippen D, Gibson BW, Bredesen DE. Identification of new modulators and protein alterations in non-apoptotic programmed cell death. Journal of Cellular Biochemistry. 2010;111(6):1401-1412. DOI: 10.1002/jcb.22870
  114. 114. Tay KH, Luan Q , Croft A, Jiang CC, Jin L, Zhang XD, et al. Sustained IRE1 and ATF6 signaling is important for survival of melanoma cells undergoing ER stress. Cellular Signalling. 2014;26(2):287-294. DOI: 10.1016/j.cellsig.2013.11.008
  115. 115. Nedungadi D, Binoy A, Pandurangan N, Pal S, Nair BG, Mishra N. 6-Shogaol induces caspase-independent paraptosis in cancer cells via proteasomal inhibition. Experimental Cell Research. 2018;364(2):243-251. DOI: 10.1016/j.yexcr.2018.02.018
  116. 116. Wang WB, Feng LX, Yue QX, Wu WY, Guan SH, Jiang BH, et al. Paraptosis accompanied by autophagy and apoptosis was induced by celastrol, a natural compound with influence on proteasome, ER stress and Hsp 90. Journal of Cellular Physiology. 2012;227(5):2196-2206. DOI: 10.1002/jcp.22956
  117. 117. Kim SH, Shin HY, Kim YS, Kang JG, Kim CS, Ihm SH, et al. Tunicamycin induces paraptosis potentiated by inhibition of BRAFV600E in FRO anaplastic thyroid carcinoma cells. Anticancer Research. 2014;34(9):4857-4868
  118. 118. Ram BM, Ramakrishna G. Endoplasmic reticulum vacuolation and unfolded protein response leading to paraptosis like cell death in cyclosporine a treated cancer cervix cells is mediated by cyclophilin B inhibition. Biochimica et Biophysica Acta. 2014;1843(11):2497-2512. DOI: 10.1016/j.bbamcr.2014.06.020
  119. 119. Kessel D, Reiners JJ Jr. Effects of combined Lysosomal and mitochondrial Photodamage in a non-small-cell Lung Cancer cell line: The role of paraptosis. Photochemistry and Photobiology. 2017;93(6):1502-1508. DOI: 10.1111/php.12805
  120. 120. Monel B, Compton AA, Bruel T, Amraoui S, Burlaud-Gaillard J, Roy N, et al. Zika virus induces massive cytoplasmic vacuolization and paraptosis-like death in infected cells. The EMBO Journal. 2017;36(12):1653-1668. DOI: 10.15252/embj.201695597
  121. 121. Korsnes MS, Espenes A, Hetland DL, Hermansen LC. Paraptosis-like cell death induced by yessotoxin. Toxicology In Vitro. 2011;25(8):1764-1770. DOI: 10.1016/j.tiv.2011.09.005
  122. 122. Baraz R, Cisterne A, Saunders PO, Hewson J, Thien M, Weiss J, et al. mTOR inhibition by everolimus in childhood acute lymphoblastic leukemia induces caspase-independent cell death. PLoS One. 2014;9(7):e102494. DOI: 10.1371/journal.pone.0102494
  123. 123. Xue J, Li R, Zhao X, Ma C, Lv X, Liu L, et al. Morusin induces paraptosis-like cell death through mitochondrial calcium overload and dysfunction in epithelial ovarian cancer. Chemico-Biological Interactions. 2018;283:59-74. DOI: 10.1016/j.cbi.2018.02.003
  124. 124. Mpakou VE, Nezis IP, Stravopodis DJ, Margaritis LH, Papassideri IS. Different modes of programmed cell death during oogenesis of the silkmoth Bombyx mori. Autophagy. 2008;4(1):97-100
  125. 125. Beg MA, Ginther OJ. Follicle selection in cattle and horses: Role of intrafollicular factors. Reproduction. 2006;132:365-377. DOI: 10.1530/rep.1.01233
  126. 126. Hatzirodos N, Hummitzsch K, Irving-Rodgers HF, Harland ML, Morris SE, Rodgers RJ. Transcriptome profiling of granulosa cells from bovine ovarian follicles during atresia. BMC Genomics. 2014;15:40. DOI: 10.1186/1471-2164-15-40

Written By

Nayeli Torres-Ramírez, Rosario Ortiz-Hernández, M. Luisa Escobar-Sánchez, Olga M. Echeverría-Martínez and Gerardo H. Vázquez-Nin

Submitted: 25 September 2018 Reviewed: 23 November 2018 Published: 15 February 2019