Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Biomedicine & Pharmacotherapy 176 (2024) 116815

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

The crosstalk between cell death and pregnancy related diseases: A


narrative review
Xiaowen Xie a, b, 1, Jiayu Liu c, 1, Jingyi Gao d, 1, Chenwei Shang a, e, Ying Jiang c, Lingyan Chen a,
Zhiwen Qian a, Lu Liu c, Danping Wu c, Yun Zhang f, *, Zhu Ru g, **, Yan Zhang c, a, ***
a
Wuxi Maternal and Child Health Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi, Jiangsu 214002, China
b
The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu
214023, China
c
Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Affiliated Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu
214002, China
d
Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
e
The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210029, China
f
Wuxi Maternal and Child Health Care Hospital, Affiliated Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China
g
Anqing Medical College Clinical Research Center, Anqing Municipal Hospital, Anqing 246003, Anhui, China

Abbreviations: AIF, apoptosis-inducing factor; AIM2, absent in melanoma 2; AKT, protein kinase B; AMP, adenosine monophosphate; AMPK, AMP-activated
protein kinase; APAF-1, apoptotic protease activator factor-1; ASC, apoptosis-associated speck-like protein containing CARD; ASK1, apoptosis signal-rgulating kinase
1; ATF, activating transcription factor; ATG, autophagy-related gene; ATP, adenosine triphosphate; BAK, BCL-2-associated k protein; BAX, BCL-2-associated X
protein; BCL-2, B cell lymphoma-2; BH3, BCL-2 homology region 3; BID, BH3-interacting domain death agonist; Bim, Bcl-2-Like Protein 11; CFLIP, cellular caspase-8
FLICE-like inhibitory protein; CHOP, C/EBP-homologous protein; CYLD, cylindromatosis; DAMPs, damage associated molecular patterns; DISC, death-inducing
signaling complex; DMT1, divalent metal transporter 1; DsDNA, double-stranded DNA; Elf2α, eukaryotic initiation factor-2α; Endo G, endonuclease G; ER, endo­
plasmic reticulum; ERK, extracellular regulated protein kinases; ERp29, endoplasmic reticulum protein 29; EVTs, extravillous trophoblasts; FADD, Fas-associating
protein with a novel death domain; FGR, fetal growth restriction; FIP200, focal adhesion kinase family interacting protein of 200 kD; FOXO3, forkhead box O3; FTL,
ferritin light chain; GCDCA, glycochenodeoxycholic acid; GDM, gestational diabetes mellitus; Glu, glutamicacid; GLUT, glucose transporter; GPX4, glutathione
peroxidase 4; GSDMD/E, gasdermin D/E; GSH, glutathione; Hcy, homocysteine; HDACs,, histone deacetylases; HIF-1α, hypoxia inducible factor-1α; HTRA, high-
temperature requirement serine protease A; ICP, intrahepatic cholestasis of pregnancy; IL-34, interleukin-34; IRE1, inositol-requiring enzyme 1; IUGR,, intra-uterine
growth restriction; JNK, c-Jun N-terminal protein kinase; LC3, microtubule-associated protein light chain 3; LncRNAs, long non-coding RNAs; LPS, lipopolysac­
charides; MLKL, mixed lineage kinase domain-like proteins; MOMP, mitochondrial outer membrane permeabilization; MTOR, mammalian target of rapamycin;
MTORC1, mammalian target of rapamycin complex 1; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor-κB; NINJ1, ninjurin 1; NLRC4,
NLR family CARD domain containing 4; NLRP3, NOD-like receptor family, pyrin domain-containing protein 3; P38 MAPK, p38 mitogen-activated protein kinases;
PAMPs, pathogen-associated molecular patterns; PI3K, phosphatidylinositol 3-kinase; Pe, phosphatidylethanolamine; PE, preeclampsia; PERK, protein kinase RNA-
like endoplasmic reticulum kinase; PLOOHs, phospholipid hydroperoxides; PPARγ, peroxisome proliferator-activated receptor γ; PUFAs, polyunsaturated fatty acids;
RIPK, receptor-interacting protein kinase; RM, recurrent miscarriage; ROS, reactive oxygen species; RPL, recurrent pregnancy loss; RSA, recurrent spontaneous
abortion; RTK, receptor tyrosine kinase; SENG, soluble endothelial glycoprotein; SETD7, SET domain containing lysine methyltransferase 7; SIRT, silent information
regulator factor 2-related enzyme; SMAC, second mitochondria-derived activator of caspases; STAPA2, spermatogenesis-associated protein 2; STAT1, signal trans­
ducer and activator of transcription 1; STEAP3, six-transmembrane epithelial antigen of prostate 3; TFR, transferrin receptor; TLR, toll-like receptors; TNFL, tumor
necrosis factor ligand; TNFR, tumor necrosis factor receptor; TRADD, tumor necrosis factor receptor type 1-associated death domain protein; TRAF, tumor necrosis
factor receptor-associated factor; TRAIL, TNF-related apoptosis-inducing ligand; TRIB3, tribbles3; TRIF, TIR domain-containing adaptor inducing interferon-β; TSC1/
2, tuberous sclerosis complex 1/2; TBID, truncated BH3-interacting domain death agonist; ULK1, unc-51-like kinase 1; UPR, unfolded protein response; VEGF,
vascular endothelial growth factor; VPS34, vacuolar protein sorting 34; XIAP, X chromosome-linked inhibitor of apoptosis protein; ZBP1, Z-conformation nucleic acid
binding protein 1; ZIKV, zika virus.
* Correspondenc to: Wuxi Maternal and Child Health Care Hospital,Wuxi School of Medicine, Jiangnan University, No. 48 Huaishu Road, Wuxi, Jiangsu 214002,
China.
** Correspondence to: Anqing Medical College Clinical Research Center, Anqing Municipal Hospital, No. 87 Tianzhushan East Road, Anqing, Anhui 246003, China.
*** Correspondence to: Department of Oncology, Wuxi Maternal and Child Health Care Hospital,Wuxi School of Medicine, Jiangnan University, No. 48 Huaishu
Road, Wuxi, Jiangsu 214002, China.
E-mail addresses: zhangyun2107@163.com (Y. Zhang), zhuru19790202@163.com (Z. Ru), fuyou2007@126.com (Y. Zhang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2024.116815
Received 26 March 2024; Received in revised form 10 May 2024; Accepted 21 May 2024
Available online 23 May 2024
0753-3322/© 2024 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

A R T I C L E I N F O A B S T R A C T

Keywords: Programmed cell death is intricately linked to various physiological phenomena such as growth, development,
Cell death and metabolism, as well as the proper function of the pancreatic β cell and the migration and invasion of
Pregnancy related disease trophoblast cells in the placenta during pregnancy. Traditional and recently identified programmed cell death
Apoptosis
include apoptosis, autophagy, pyroptosis, necroptosis, and ferroptosis. In addition to cancer and degenerative
Autophagy
Pyroptosis
diseases, abnormal activation of cell death has also been implicated in pregnancy related diseases like pre­
Necroptosis eclampsia, gestational diabetes mellitus, intrahepatic cholestasis of pregnancy, fetal growth restriction, and
Ferroptosis recurrent miscarriage. Excessive or insufficient cell death and pregnancy related diseases may be mutually
determined, ultimately resulting in adverse pregnancy outcomes. In this review, we systematically describe the
characteristics and mechanisms underlying several types of cell death and their roles in pregnancy related dis­
eases. Moreover, we discuss potential therapeutic strategies that target cell death signaling pathways for preg­
nancy related diseases, hoping that more meaningful treatments will be applied in clinical practice in the future.

1. Introduction interventions. To explore the roles of diverse cell death in pregnancy


related diseases, we collated a large amount of literature and found that
Cell death refers to irreversible changes such as metabolic arrest, apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis are the
structural destruction, and functional deterioration following severe main cell death modes affecting pregnancy related diseases. Based on
cellular damage. Formerly perceived as stemming from either apoptosis the keywords of five cell death and pregnancy related diseases, we
or necrosis, recent discoveries have unveiled several novel modes of cell conducted a thorough search of recent literature, filtering out works of
death, each with distinct pathways [1]. Different types of cell death significant relevance and high quality. Subsequently, we meticulously
exhibit diverse characteristics. For instance, as an enzyme-dependent analyzed, organized, and summarized these findings (Fig. 1A). In this
biochemical process, apoptosis performs in a controlled manner, with review, we centered on these five cell death modes that have been
cellular contents mostly removed by tissue-resident professional deeply implicated in pregnancy related diseases. We elaborated their
phagocytes or neighboring non-professional phagocytes instead of being respective mechanisms of action that might induce pregnancy related
spilled over into the extracellular milieu [2]. On the contrary, necrosis is diseases, particularly PE, ICP, and GDM. The impacts of these cell death
commonly described as uncontrolled cell death, typically accompanied modes on pregnancy related diseases were also described. At the same
by a severe invasion, leading to linkage of cellular contents and subse­ time, we summarized multiple ways in which cell death can exert pos­
quent impairment [1]. Interestingly, necroptosis displays the features of itive effects or inhibit the deterioration of pregnancy related diseases,
necrosis but is tightly regulated, resembling a highly regulated form of presenting several cell death-related treatment approaches that may
necrosis [3]. transform the therapeutic landscape (Fig. 1B).
The dysregulation of cell death has been implicated in a range of
diseases. Overactivation of cell death increases the susceptibility to 2. Apoptosis
neurodegenerative diseases, whereas deficits in cell death are associated
with tumors and autoimmune diseases [4–6]. The placenta serves as a 2.1. Mechanisms of apoptosis
bridge between mother and fetus, facilitating material exchange, hor­
monal regulation, defense against pathogens, and immune tolerance Apoptosis refers to the self-regulated and systematic cell death
maintenance. These types of cell death also exert pivotal roles in both controlled by genes to maintain a stable internal environment. In 1972,
the physiological development and abnormal disorders of the placenta Kerr, Wyllie, and Currie first proposed the concept of “apoptosis,”
and the fetus. Apoptosis contributes to the placenta’s growth through describing its morphological features as cellular shrinkage, membrane
providing efficient utero-placental blood perfusion to the early fetus [7]. blebbing, nuclear fragmentation, chromatin condensation, and the for­
The number of apoptotic cells in the placenta fluctuates throughout mation of apoptotic body [14]. In general, apoptosis can be triggered
gestational stages. Apoptosis in the placenta mostly occurs in syncytio­ through three pathways: the intrinsic pathway, the extrinsic pathway,
trophoblasts, which act as a barrier between fetal and maternal circu­ and the endoplasmic reticulum stress (ER stress)-induced pathway
lation and are essential for maintaining placental homeostasis [8]. (Fig. 2).
Intrahepatic cholestasis of pregnancy (ICP) results in elevated bile acid The intrinsic pathway, also known as the mitochondrial pathway of
levels and prolonged exposure to toxic bile acids, thereby compromising apoptosis, is initiated by intracellular stressors [15]. Once the cell per­
trophoblast cell function and impairing autophagic processes. [9]. ceives these stressors, mitochondrial outer membrane permeabilization
Furthermore, increased placental size and mass in gestational diabetes (MOMP) will occur, leading to cytochrome c releasing from the inter­
mellitus (GDM) correlate with a considerable reduction in trophoblast membrane space of mitochondria into the cytosol [16]. In this process,
apoptosis [10,11]. Nutritional deficiencies during pregnancy may proapoptotic proteins of the B cell lymphoma-2 (BCL-2) family, promi­
weaken defenses against environmental pollutants and precipitate nently BCL-2-associated X protein (BAX) and BCL-2-associated k protein
changes in the body’s internal environment, fostering apoptosis, ne­ (BAK), and dynamin-related protein 1-induced cristae remodeling play a
crosis, and other aberrant cell deaths, leading to abnormal invasion of crucial role [17,18]. At the same time, BCL-2 homology region 3
the trophoblast and the development of preeclampsia (PE) [12]. In (BH3)-only proteins directly target BAX/BAK or deactivate
addition, placental cells are extremely sensitive to ferroptosis. Factors anti-apoptotic BCL-2 proteins to trigger apoptosis [19]. Subsequently,
such as hypoxia or reactive oxygen species (ROS) injury can all indi­ cytochrome c attaches to apoptotic protease activator factor-1 (APAF-1)
rectly induce placental ferroptosis [13]. monomers, prompting the conformational shift in APAF-1 and its
Previous reviews have tended to focus on the connections between interaction with procaspase-9, thus forming the apoptosome, where
cell death and cancer and degenerative diseases, or single cell death in activated caspase-9 cleaves the precursors of caspase-3 and -7 and un­
pregnancy related diseases, but there are few comprehensive reviews of leashes their apoptotic executioner function [20,21]. X
the relationship between various forms of cell death and pregnancy chromosome-linked inhibitor of apoptosis protein (XIAP) prevents pro­
related diseases. Our review aims to fill the gap in this field and provide cessed caspase activity to elevate the threshold for apoptosis activation
reference and guidance for both basic research and clinical [22]. However, second mitochondria-derived activator of caspases

2
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

(SMAC) and high-temperature requirement serine protease A2 (HTRA2), transcription factor 6 (ATF6), all of which can induce apoptosis through
co-released with cytochrome c during MOMP, serve as XIAP inhibitors activating C/EBP-homologous protein (CHOP) [28,29]. In short, the
[23]. equilibrium between pro-apoptotic and anti-apoptotic factors ensures
The extrinsic pathway, also called the death receptor pathway of the normal progress of apoptosis.
apoptosis, begins with the binding of death ligands to death receptors
[24]. Once the combination happens, adapter proteins tumor necrosis
factor receptor type 1-associated death domain protein (TRADD) or 2.2. Apoptosis in pregnancy related diseases
Fas-associating protein with a novel death domain (FADD) and initiator
procaspases (procaspase-8 and -10) will be recruited, resulting in the 2.2.1. Apoptosis in PE
assembly of the death-inducing signaling complex (DISC) [25]. In Type I PE, one of the most fearful complications of pregnancy, frequently
cells, activated caspase-8 directly activates the executioner caspases, manifests as new-onset hypertension, proteinuria, or other end-organ
while in Type II cells, caspase-8 cleaves the BH3-interacting domain damage during the third trimester, threatening the life safety of both
death agonist (BID) to truncated BID (tBID), which enters mitochondria mother and fetus [30]. Although the precise pathogenesis remains
and activates the intrinsic apoptotic pathway [26]. Meanwhile, the incompletely elucidated, it is known that abnormal spiral artery
anti-apoptotic protein cellular caspase-8 (FLICE)-like inhibitory protein remodeling and inadequate trophoblast invasion are closely related to
(cFLIP) can suppress the activity of caspase-8 [27]. PE [31]. In addition, inflammation and increased apoptosis in villous
Endoplasmic reticulum secretory failure, infections, hypoxia, and cytotrophoblasts during early pregnancy augment the risk of PE [32]. In
several chemicals can all trigger the buildup of unfolded or misfolded the placenta of patients with PE, highly expressed IFN-γ upregulates the
proteins, which induces ER stress-induced cell death [16]. Unfolded phosphorylation levels of Janus kinase 1 and signal transducer and
protein response (UPR) pathway, initiated by protein misfolding, has activator of transcription 1 (STAT1) to inhibit the migration and inva­
three main ingredients: inositol-requiring enzyme 1 (IRE1), protein ki­ sion of trophoblast cells while promoting apoptosis [33]. Furthermore,
nase RNA-like endoplasmic reticulum kinase (PERK), and activating oxidative stress is associated with the pathogenesis of PE. Increased
sensitivity to apoptosis has been observed in chorionic trophoblast cells

Fig. 1. A brief overview of cell death and pregnancy related diseasesA. From the articles we obtained and screened, we summarized the five modes of cell death most
closely related to pregnancy related diseases, which may be involved in adverse pregnancy outcomes. B. Stressors present in the environment and in the body of
pregnant women lead to the dysregulation of cell death in some tissues and organs, thereby becoming a potential cause of pregnancy related diseases. Meanwhile,
pregnancy related diseases can further exacerbate the dysregulation of cell death.

3
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

in the placentas of pregnant women with intra-uterine growth restric­ the miR-128–3p/BCL2 Interacting Protein 3/mammalian target of
tion (IUGR) and PE, which makes trophoblast cells more susceptible to rapamycin (mTOR) signaling pathway to inhibit trophoblast cell pro­
oxidative damage [34,35]. Evidence suggests that excessive placental liferation and facilitate cell apoptosis in PE [42].
ROS activate p38 MAPK and nuclear factor-κB (NF-κB) pathways,
incurring trophoblast apoptosis [36]. As a nicotinamide adenine 2.2.2. Apoptosis in intrahepatic cholestasis of pregnancy
dinucleotide-dependent histone deacetylase, SIRT1 controls apoptosis ICP presents with pruritus, right upper quadrant pain, sleep depri­
and autophagy via deacetylating its downstream targets. Elevated vation, and increased serum bile acids and liver transaminases during
intracellular ROS levels weaken the activity of SIRT1 in PE placentas, the second and third trimesters, related to preterm birth, respiratory
resulting in dysregulation of apoptosis [37]. Moreover, studies have distress syndrome, and fetal demise [43]. Evidence indicates that
demonstrated that an imbalance in micronutrients such as folate and placental apoptosis induced by high concentrations of bile acids may be
vitamin B12 causes oxidative stress and higher homocysteine levels, the major pathological event of ICP and injure the structure and function
which gives rise to increased placental apoptosis and changes in of the placenta, ultimately leading to undesirable fetal outcomes [44].
placental angiogenesis [38]. The complement system is fundamental to MiRNAs are small noncoding RNA molecules that inhibit the expression
the clearance of apoptotic cells, but the reaction can be inflammatory in of downstream genes by binding to the target mRNA and are implicated
PE, increasing the risk of pregnancy complications and impaired endo­ in various human diseases [45]. According to recent studies, several
thelial integrity [39]. What’s more, dysregulation of certain genes or exosome miRNAs may serve as novel biomarkers to improve the diag­
long non-coding RNAs (lncRNAs) also has an impact on trophoblast nosis and prognosis of ICP [46]. Additionally, it is speculated that
invasion and apoptosis [40,41]. For example, lncRNA BNIP3P1 targets miR-221/222 may increase apoptosis in HTR-8 cells through

Fig. 2. Apoptosis signaling pathway in pregnancy related diseases.The main components of apoptosis include the intrinsic pathway, the extrinsic pathway, and the
ER stress-induced pathway. In the intrinsic pathway, intracellular stressors such as DNA damage and toxic agents activate BAX and BAK, and then boost MOMP. This
process results in the release of cytochrome c and the formation of the apoptosome, subsequently activating caspase-9 and initiating the caspase-processing cascade.
The extrinsic pathway is triggered by the interaction between death ligands and death receptors, which contributes to the formation of DISC and caspase-8 activation,
thereby initiating an amplifying cascade. ER stress also mediates apoptosis through multiple pathways. Several stressors (i.e., ROS, some microRNAs, nutrient
deprivation, and environmental pollutants) associated with pregnancy related diseases participate in apoptosis at different sites, leading to the occurrence of diseases
and promoting disease progression.(AIF, apoptosis-inducing factor; Endo G, endonuclease G; Bim, Bcl-2-Like Protein 11; Hcy, homocysteine; TNFL, tumor necrosis
factor ligand; TNFR, tumor necrosis factor receptor; TRAIL, TNF-related apoptosis-inducing ligand; SIRT1, silent information regulator factor 2-related enzyme 1;
ASK1, apoptosis signal-rgulating kinase 1; JNK, c-Jun N-terminal protein kinase; p38 MAPK, p38 mitogen-activated protein kinases; elf2α, eukaryotic initiation
factor-2α; ATF4, activating transcription factor 4; TRIB3, tribbles3; AKT, protein kinase B).

4
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

downregulating the expression of BCL-2 [47]. Has_circ_0060731 maintenance [69]. Studies indicate that disruption of
potentially modulates trophoblast cell apoptosis via the macrophage-induced, FasL-mediated apoptosis may contribute to RM
miR-21–5p-PDCD4/ESR1 axis [48]. Endoplasmic reticulum protein 29 [70]. Histone deacetylases (HDACs), known as histone modification
(ERp29) influences the biosynthesis of various secretory proteins as well enzymes, are crucial for the differentiation, activation, and apoptosis of
as apoptosis and oxidative stress regulation [44,49]. Data show that various immune cells, including macrophages. Knockdown of HDAC8 in
overexpression of ERp29 induces apoptosis in response to elevated bile decidual macrophages from RM patients results in suppressed M2 acti­
acid concentrations in vitro [50]. Furthermore, ER stress-mediated vation, and differentiated THP-1 macrophages are more likely to un­
trophoblast apoptosis may have a crucial role in the pathogenesis of dergo apoptosis through activating the ERK pathway [71].
ICP, as a study has demonstrated that deoxycholic acid can induce
apoptosis by promoting the expression of mRNA and proteins encoded 3. Autophagy
by ER stress-related genes in trophoblasts in a concentration-dependent
manner [51]. Animal experiments have also shown severe fetal 3.1. Mechanisms of autophagy
myocardial structure damage and increased apoptosis in ICP patients
[52]. Autophagy is a process of degradation and regeneration in cells,
which encapsulates damaged or excess cellular components into vesicles
2.2.3. Apoptosis in GDM and decomposes them for reuse, akin to fallen petals transforming into
GDM is a typical pregnancy complication characterized by abnormal soil to nourish the flower [72]. Autophagy can induce cell death by
maternal glucose metabolism and is concerned with higher risks of initiating apoptosis or through independent pathways [73].
preterm birth, fetal macrosomia, and neonatal respiratory distress syn­ Several stresses, including oxidative stress, nutritional scarcity, and
drome [53]. Neutrophil extracellular traps triggered by hyperglycemia UPR, can trigger the self-protection mechanism of autophagy [8]. The
cause trophoblast apoptosis through the generation of intracellular ROS signaling pathways are determined by several autophagy-associated
and downregulation of extracellular regulated protein kinases (ERK) proteins. Unc-51-like kinase 1 (ULK1), focal adhesion kinase family
pathway [54]. However, another study has found that GDM reduces interacting protein of 200 kD (FIP200), autophagy-related gene 13
apoptosis of the placenta in term [10]. Additionally, lncRNAs have a (ATG13), and ATG101 collaborate to form a preinitiation complex and
place in GDM, similar to their involvement in PE and ICP. By targeting initiate this pathway. The activity of the ULK complex is directly regu­
specific cellular molecules and signaling pathways, certain up-regulated lated by the mammalian target of rapamycin complex 1 (mTORC1) and
miRNAs in GDM patients can attenuate cell viability, reduce prolifera­ AMP-activated protein kinase (AMPK) [74]. mTOR, a highly conserved
tion, and promote the apoptosis of pancreatic β cells, thereby compro­ mitogen-activated protein kinase, binds with multiple companion pro­
mising pancreatic β cell function [55–57]. Several cardio-protective teins to form distinct signaling complexes, such as mTORC1. mTORC1
miRNAs which prevent cardiomyocyte apoptosis are revealed to be inhibits the activity of ULK complexes by phosphorylation and regulates
upregulated in serum from GDM during the first trimester of pregnancy the expression and function of other key proteins for autophagy, thereby
but considerably suppressed in the third trimester [58]. Besides, it has restraining autophagy [75]. By contrast, triggered by ATP depletion or
been noted that GDM and inflammation have a substantial relationship. metabolic stress, AMPK encourages the initiation of autophagy through
According to studies, interleukin-34 (IL-34) accelerates pancreatic direct and indirect pathways [76]. Therefore, mTOR can be negatively
apoptosis in GDM patients by encouraging STAT3 phosphorylation regulated to promote autophagy, while AMPK plays an antagonistic role
through colony-stimulating factor 1 receptor, and it may be secreted into against mTOR. Afterwards, the vacuolar protein sorting 34 (VPS34)
the fetal circulation, influencing fetus metabolism [59]. complex, activated by the ULK complex, cooperates with the ULK
complex to promote the creation of autophagosomes [77]. Finally,
2.2.4. Apoptosis in fetal growth restriction autophagosomes combine with lysosomes to form autolysosomes, where
Fetal growth restriction (FGR), also known as IUGR, refers to a the contents are degraded by proteases derived from lysosomes (Fig. 3)
sonographic estimated fetal weight below the 10th percentile or the [1].
abdominal circumference below the 10th percentile, causing secondary
diseases in adulthood and an increased risk of metabolic syndrome [60]. 3.2. Autophagy in pregnancy related diseases
Due to increased syncytiotrophoblast apoptosis, the placenta in FGR
exhibits both damage and healing, along with villus dysplasia [61]. 3.2.1. Autophagy in PE
Environmental pollutants, such as heavy metal cadmium, can build up in The pathogenesis of PE remains poorly understood, but many studies
the body, resulting in placental apoptosis and FGR through the degra­ have indicated that the development of PE is related to placental
dation of myeloid cell leukemia-1 mediated by mitochondrial ROS [62]. dysfunction [78]. Under hypoxic conditions, although the standard of
MiRNA such as miR-1227–3p is downregulated in the FGR patients’ cell viability and proliferation remain similar, the depth of invasion of
placentas, yet it may contribute to tumor formation by preventing extravillous trophoblasts (EVTs) cell lines is significantly shallower
apoptosis and boosting cell proliferation and survival [63]. In addition, compared to normal, which may be attributed to the inhibition of
the placenta-specific 1 gene related to pregnancy and placental devel­ autophagy by soluble endothelial glycoprotein (sENG) in EVTs [79]. At
opment and antiapoptotic BCL-2 gene present reduced expression in the same time, activated transcription factor hypoxia inducible factor-1α
FGR tissues, diminishing their capacity to inhibit trophoblast cell (HIF-1α) promotes the production of ROS, stimulating the placenta to
apoptosis at low oxygen concentrations [64,65]. release sENG, which further hinders the formation of blood vessels and
aggravates placental hypoxia, thus exacerbating placental dysfunction
2.2.5. Apoptosis in recurrent miscarriage [80,81]. A study demonstrated that activation of autophagy in placental
The definition of recurrent miscarriage (RM) is controversial, but it is trophoblasts prevents bisphenol A-induced apoptosis and mitochondrial
evident that RM correlates with long-term health problems extending dysfunction, reducing the risk of pregnancy-related diseases including
beyond pregnancy, and one of the primary factors of RM is trophoblast PE [82]. Furthermore, through deacetylating several substrates, SIRT1
function dysregulation [66]. High levels of sprouty 4 may upregulate may promote trophoblast autophagy, thereby providing protection
IFN-γ/phosphatidylinositol 3-kinase (PI3K)/AKT-induced STAT1 acti­ against PE [83]. However, excessive autophagy induced by long-term
vation, contributing to the onset and advancement of RM [67]. In hypoxia can disrupt trophoblast cell invasion and vascular remodel­
first-trimester RM patients, cyclin A2 downregulation affects the cell ing, impairing placental function [84]. Autophagy activation mediated
cycle and inhibits HTR8 apoptosis via the p53 pathway [68]. Moreover, by ceramide and hydrolase activity induced by oxidative stress impairs
macrophages are typically regarded as necessary for pregnancy placental function in PE patients [85]. AMPKα phosphorylation

5
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

Fig. 3. Autophagy signaling pathway in pregnancy related diseases.Growth factors like insulin and VEGF bind to RTKs on cell membranes, activating the PI3K-AKT-
mTOR pathway, which promotes the absorption and utilization of nutrients such as glucose. Deficiencies in nutrition, ATP, and oxygen activate AMPK, which in turn
activates ULK1 through direct phosphorylation or inhibiting mTOR. The ULK complex works with VPS34 complex to promote phagophore transforming into
autophagosomes under the regulation of Atg5-Atg12-Atg16L and LC3-II. Finally, autophagosomes merge with lysosomes to digest and break down the inner contents.
Pregnancy related diseases are often accompanied by autophagy disorders, such as hyperactivation of autophagy induced by trophoblast cell hypoxia. Overactivation
of autophagy can disrupt cellular metabolic regulation, leading to the onset of diseases.(VEGF, vascular endothelial growth factor; RTK, receptor tyrosine kinase;
AMP, adenosine monophosphate; ATP, adenosine triphosphate; TSC1/2, tuberous sclerosis complex 1/2; LC3, microtubule-associated protein light chain 3; pe,
phosphatidylethanolamine).

decreases the expression of peroxisome proliferator-activated receptor γ breakdown of autophagosomes [88]. As a result, autophagy fails to exert
(PPARγ), which is instrumental in regulating spiral artery development its protective role, disrupting cellular homeostasis and further contrib­
and placental function and is associated with the onset of PE [84,86]. In uting to hepatocyte damage in cholestasis. Research has found that bile
conclusion, autophagy is a double-edged sword for PE. In the hypoxic acids exert different effects on autophagy under different circumstances.
environment, autophagy plays a pivotal role in early placental growth Glycochenodeoxycholate was once believed to induce autophagy in
and development, whereas insufficient or unduly activation of auto­ hepatocellular carcinoma, while glycochenodeoxycholic acid (GCDCA)
phagy can compromise the function of placenta. has been discovered to inhibit the formation of autophagosomes and
impair lysosomal function by controlling lysosomal proteolysis and
3.2.2. Autophagy in ICP increasing lysosomal pH in cholestasis [89,90]. Transcription factor E3,
Patients with ICP frequently have elevated levels of bile acids, which belonging to the same family of transcription factors as transcription
impede autophagy in trophoblasts [9]. Bile acids activate the nuclear factor EB and interacting with transcription factor EB in the regulation
receptor farnesoid X receptor, which is predominantly expressed in the of autophagy and lysosomal biogenesis, experiences signaling disruption
liver and is thought to inhibit autophagy, resulting in decreased auto­ by GCDCA [91]. Consequently, abnormalities in autophagy activity
phagy activity in hepatocytes [87]. Farnesoid X receptor-dependent caused by the accumulation of GCDCA detrimentally affects
Rubicon, an inhibitor of autophagosome maturation, interferes with hepatocytes.
the combination of autophagosome and lysosomal, hindering the proper

6
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

3.2.3. Autophagy in GDM regulation of ULK1 by miR-33b-3p, hence contributing to placental


The body undergoes a number of changes during pregnancy, autophagy in GDM [96]. In summary, hyperglycemia can both over
including a rise in hormonal and blood glucose levels to support fetal activate and inhibit autophagy, and the effects on placental function
growth. Increased hormone levels in pregnant women, such as cortisol, require deeper investigation.
progesterone, and other antagonistic insulin-like substances, may
induce insulin resistance, leading to reduced glucose uptake by cells and 3.2.4. Autophagy in other pregnancy related diseases
consequently higher blood sugar levels [92]. Normally, islet β cells can In addition, autophagy may be linked to other diseases, such as FGR
maintain blood glucose balance through autophagy, while hyperglyce­ and RM. Deterioration of the endometrial environment is an important
mia can induce excessive autophagy of pancreatic β cells and worsen trigger of RM. Aberrant crosstalk between amino acid metabolism and
insulin resistance [93]. By raising advanced glycation end products or autophagy may elevate autophagic activity, thus impairing the endo­
oxygen radical levels in cells, hyperglycemia promotes ROS and oxida­ metrial microenvironment and inducing RM [97]. Cadmium triggers
tive stress. As a result, it induces excessive autophagy of trophoblasts, BCL2 interacting protein 3-dependent mitophagy in placental tropho­
impedes placental angiogenesis, and causes placental ischemia and blasts and reduces maternal serum progesterone levels, resulting in FGR
hypoxia [94]. At the same time, it was found in in vitro cell culture [98]. Autophagy serves as a protective mechanism for trophoblast cells
experiments that hyperglycemia would induce excessive autophagy in against oxidative stress, while overactivity of autophagy may ultimately
trophoblasts. Compared with cells cultured in a normal sugar environ­ compromise the function of trophoblasts, resulting in placental hypo­
ment, trophoblasts exposed to high glucose levels showed a significant perfusion, which in turn causes an insufficient supply of both nutrients
increase in the expression of the autophagy marker LC3 [94]. However, and oxygen in fetal development [99].
another research has proved that hyperglycemia can increase
low-density lipoprotein transcytosis by suppressing autophagy in human
endothelial cells [95]. Bao Y et al. revealed several dysregulated circular
RNAs in GDM placenta that influenced the PI3k-Akt and insulin
signaling pathways. For example, increased circCDH2 may inhibit the

Fig. 4. Pyroptosis signaling pathway in pregnancy related diseases.In the canonical pathway, a range of external and internal stressors recruit effector proteins
(mainly ASC and NLRP3) to assemble inflammasomes, which in turn activate caspase-1. In the non-canonical pathway, LPS activates caspase-4, -5, and -11, which
cleave GSDMD into GSDMD-C and GSDMD-N. The latter oligomerizes and forms pores in plasma membranes, resulting in the influx of water and sodium ions and the
efflux of DAMPs, leading to membrane rupture. At the same time, caspase-1 proteolytically matures proIL-18 and proIL-1β into IL-18 and IL-1β, which spill
extracellularly and mediate the inflammatory response. Moreover, activated caspase-3 cleaves GSDME and promotes pore formation. Multiple stressors and mo­
lecular dysregulation during pregnancy can promote the formation of inflammasomes, induce placental inflammatory response and pyroptosis, and ultimately lead to
adverse pregnancy outcomes.(DAMPs, damage associated molecular patterns; PAMPs, pathogen-associated molecular patterns; dsDNA, double-stranded DNA; AIM2,
absent in melanoma 2; NLRC4, NLR family CARD domain containing 4; NINJ1, ninjurin 1; ZIKV, zika virus; FOXO3, forkhead box O3).

7
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

4. Pyroptosis elevates thioredoxin-interacting protein and then activates the NLRP3


inflammasome, boosting the production of active caspase-1 [122]. In
4.1. Mechanisms of pyroptosis addition, glycolysis in the trophoblast layer of PE patients is upregulated
but less efficient, resulting in attenuated ATP synthesis, potentially
Pyroptosis is a recently discovered mode of programmed cell death associated with LPS-induced toll-like receptors 4 (TLR4)/NF-κB activa­
marked by its reliance on inflammatory cysteine asparaginases (mainly tion that disrupts the mitochondrial structure and function of tropho­
caspase-1, -4, -5, and -11) and the release of numerous pro-inflammatory blasts and induces pyroptosis [123].
molecules [100]. Pyroptosis related pathways are categorized into ca­
nonical and non-canonical pathways (Fig. 4). 4.2.2. Pyroptosis in other pregnancy related diseases
The canonical pathway is mediated by inflammasome assembly with Pyroptosis is implicated in placental implantation, formation, and
gasdermin D (GSDMD) cleavage and the release of IL-1β and IL-18 [101, anti-infection; nonetheless, pyroptosis hyperactivation can be mutually
102]. Upon the host’s resistance to microbial infection or other stressors, causal with obstetric complications [124]. Recent research has revealed
the multimolecular complex inflammasomes are activated, promoting that maternal hypothyroidism activates classical inflammatory and
an adaptive immune response. Inflammasome assembly begins with pyroptosis pathways at the maternal-fetal interface by triggering
cytoplasmic pattern recognition receptors (PRR), and the majority of oxidative stress and ER stress [125]. ZIKV has been shown to cause
inflammasomes contain the caspase recruitment domain of the adapter placental pyroptosis through activating the actuator GSDME in vitro and
apoptosis-associated speck-like protein containing CARD (ASC), as well vivo, leading to adverse fetal outcomes in ZIKV-infected mice. In
as procaspase-1 [103–106]. In the inflammasome, activated caspase-1 mechanisms, the recognition of viral genomic RNA by retinoic
cleaves GSDMD into GSDMD-CT and GSDMD-NT, and the latter perfo­ acid-inducible gene I induces the release of TNF-α, which activates
rates the cell membrane to create a non-selective pore, leading to cell caspase-8 and caspase-3 and leads to the cleavage of GSDME, associated
swelling and pyroptosis [107–109]. In addition, caspase-1 cleaves the with the creation of a dysplastic fetus [126]. Pregnant women with GDM
precursors of IL-1β and IL-18 into their active forms, which are released are frequently accompanied by hyperglycemia. It has been revealed that
in the extracellular space and induce an inflammatory response and exposure of TP53-induced glycolysis and apoptosis regulator-deficient
pyroptosis [110]. trophoblasts to high glucose induces the assembly of the NLRP3
In the non-canonical pathway, cytoplasmic lipopolysaccharides inflammasome and leads to pyroptosis [127]. TP53-induced glycolysis
(LPS) activate caspase-4, -5, and -11, initiating pyroptosis via cleaving and apoptosis regulator deficiency is followed by overproduction and
GSDMD into GSDMD-NT, which aggregates and translocates to the cell release of ROS, along with poor placental decidualization and adverse
membrane to form a pore [111]. Cleavage of GSDMD causes the fetal outcomes [128]. Recurrent spontaneous abortion (RSA) is a com­
exocytosis of K+, inducing the assembly of the NOD-like receptor fam­ mon pregnancy complication that significantly impacts the physical and
ily, pyrin domain-containing protein 3 (NLRP3) inflammatory vesicle, mental health of pregnant women. Although the mechanisms behind
ultimately leading to pyroptosis [112]. RSA remain unclear, the high mobility group box 1 molecule has been
Besides these two main pathways, specific stimuli can lead to found to be highly expressed at the maternal-fetal interface in patients
caspase-3 activation and induce the cleavage of gasdermin E (GSDME) to with unexplained RSA [129]. High expression of high mobility group
GSDME-NT, which also participates in the formation of plasma mem­ box 1 in RSA group is actively phagocytosed by macrophages, which
brane pores, eventually resulting in pyroptosis [113]. However, recent activates pyroptosis via the TLR2/TLR4/NF-kB pathway, leading to
studies have shown that GSDMD-mediated pyroptosis can be hindered inflammation and ultimately triggering RSA [130].
by certain factors. Cleavage of GSDMD by caspase-3 and -7 at the Asp87
site leads to inactivation of GSDMD’s pyroptosis activity [114]. Addi­ 5. Necroptosis
tionally, the endosomal sorting complex required for the transport
mechanism eliminates the GSDMD pore at the plasma membrane, 5.1. Mechanisms of necroptosis
thereby inhibiting GSDMD-mediated pyroptosis and restricting the
release of IL-1β [115]. Recently, it has been reported that activated As a regulated form of necrotic cell death distinct from conventional
p-STAT3 promotes nuclear PD-L1 translocation under hypoxia. Nuclear apoptosis signaling pathways, necroptosis is independent of caspase
PD-L1 and p-STAT3 synergistically enhance GSDMC expression, and activity and exhibits necrotic phenotypes, including organelle swelling
activated caspase-8 can cleave GSDMC into GSDMC-NT, ultimately and cytolysis [131]. Necroptosis can be activated by the same triggers as
leading to pyroptosis [116]. apoptosis and serves as a backup mechanism for apoptosis [132].
Necroptosis is initiated by various cytokines and PRRs. Key proteins
4.2. Pyroptosis in pregnancy-related diseases involved in executing necroptosis include receptor-interacting protein
kinase 1(RIPK1), RIPK3 and mixed lineage kinase domain-like proteins
4.2.1. Pyroptosis in PE (MLKL), which combine to form the necrosome [133]. Upon activation
Pyroptosis has a widespread impact on obstetric disorders, particu­ of TNF signaling, RIP1 undergoes deubiquitylation by cylindromatosis
larly PE. Compared to healthy pregnant women, pregnant women with (CYLD) and interacts with RIPK3 in the absence of caspase-8 activity
PE exhibit increased levels of NLRP3 inflammasome expression in both [24]. In addition, TLR3 binding to dsDNA, TLR4 attaching to LPS, and
blood cells and the placenta [117]. The secretion of multiple inflam­ Z-conformation nucleic acid binding protein 1 (ZBP1) detecting DNA
matory mediators triggered by persistent inflammation in endothelial viruses can all activate RIPK3 [134,135]. RIPK3 then phosphorylates
cells leads to activation of NLRP3, which causes release of IL-1β and MLKL, prompting MLKL to oligomerize and transfer to the plasma
IL-18 through the pores formed by the GSDMD, resulting in focal cell membrane. Consequently, there is an increase in plasma membrane
death and then triggering PE [118,119]. In placental tissues from PE permeability, resulting in uncontrolled release of intracellular contents
patients and in hypoxia/reoxygenation-treated HTR8/Svneo cells, and eventual cell death (Fig. 5) [136]. Besides, there exist other
heightened expressions of SET domain containing lysine methyl­ signaling pathways that mediate necroptosis, and more details warrant
transferase 7 (SETD7) and HTRA1 propel pyroptosis and impair cell further investigation.
migration and invasion [120]. Down-regulated HDAC2 in PE patients Necroptosis mediators have been linked to the pathophysiology of
accompanies elevated levels of FOXO3 and PERK, as well as IL-1β and autoimmune disorders, acute respiratory distress syndrome, myocardial
IL-18 expression, inhibiting cell proliferation, migration, and invasion, infarction, stroke, and other diseases. The modulatory effects of nec­
ultimately leading to trophectodermal pyroptosis [121]. Moreover, roptosis on the immune system and the underlying link between nec­
excessive ER stress induced by hypoxia in PE hyperactivates UPR, which roptosis and several other forms of programmed cell death, as well as

8
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

Fig. 5. Necroptosis signaling pathway in pregnancy related diseases.TNF-α binding to TNFR1 induces the formation of complex I. In conditions where c-FILP levels
are low and caspase-8 maintains normal activity, apoptosis is initiated. By contrast, when caspase-8 is inhibited, RIPK1 participates in the formation of complex IIb
under the deubiquitylation of CYLD and activates RIP3. Subsequently, RIP3 phosphorylates MLKL in the necrosome, causing MLKL oligomerization and translocation
to the plasma membrane, compromising the integrity of the membrane, and resulting in the influx of sodium ions and the efflux of DAMPs. Additionally, LPS, viruses,
and ER stress can exacerbate the detrimental effect of MLKL on the plasma membrane via different pathways. Several important components of necroptosis are
significantly elevated in models of pregnancy-related diseases, suggesting their crucial role in disease pathogenesis.(TRAF, tumor necrosis factor receptor-associated
factor; STAPA2, spermatogenesis-associated protein 2; TRIF, TIR domain-containing adaptor inducing interferon-β).

various inhibitors targeting the regulatory elements of the necroptotic promotes necroptosis and inhibits trophoblast invasion and migration in
process, are currently under development [137,138]. hypoxia [143]. Consequently, cell invasion stopping at a superficial
level impedes the formation of new blood vessels, causing placental
hypoxia and dysfunction. What’s more, inhibition of G protein-coupled
5.2. Necroptosis in pregnancy related diseases receptor kinase 2 in the PE placenta contributes to inadequate vascular
remodeling and enhanced trophoblast necroptosis with the activation of
5.2.1. Necroptosis in PE RIPK1, RIPK3, and MLKL [144]. According to a recent study based on
Current research suggests that the early placenta contains important necroptosis-related differentially expressed genes, necroptosis may
elements of necroptosis, with emerging data indicating a close associa­ encourage the progression of PE through inducing immunological
tion between necroptosis and the onset of PE [139]. The formation of the infiltration and immune responses [145]. In summary, various func­
RIPK1-RIPK3 complex depends on the NAD-dependent deacetylase tional proteins and immune factors regulate necroptosis in the placenta,
SIRT2, which deacetylates and activates RIPK1 [140]. SIRT2 has been thus causing placental dysfunction in different ways.
found in various placental components, including the amnion epithe­
lium, decidual cells, cytotrophoblasts, and vascular endothelial cells 5.2.2. Necroptosis in other pregnancy related diseases
[141]. A large amount of SIRT2 in the placenta may trigger the creation Necroptosis is implicated in other pregnancy related diseases such as
of the RIPK1-RIPK3 complex and send death signals, potentially trig­ RM and FGR. According to animal experiments, deficiency of PR-SET7 in
gering necroptosis and shedding of trophoblast cells. Elevated levels of the trophoblast inhibits endogenous retroviruses, which causes an
these exfoliated aggregates of trophoblast cells have been found in the abnormal buildup of double-stranded RNA in the cytoplasm, giving rise
circulatory system of PE patients, which can cause placental endothelial to inflammation, necroptosis, and ultimately miscarriage [146,147].
dysfunction and induce PE [142]. In addition, Yu H et al. discovered that Currently, there are few studies on the relationship between necroptosis
the decrease in the expression of SIRT3 in the trophoblast of PE patients and pregnancy related diseases, and more underlying mechanisms still
led to increased expression of RIPK1, RIPK3, and p-MLKL, which

9
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

need to be further studied. (DMT1) [149]. However, this process intensifies with increased
expression of the transferrin receptor, leading to ferroptosis. Free Fe2+
6. Ferroptosis serves as a catalyst for the generation of •OH and hydroxide ions from
H₂O₂, resulting in the peroxidation of PUFAs such as arachidonic acid
6.1. Mechanisms of ferroptosis and adrenic acid [152]. Nevertheless, the exact cellular membranes
related to the lipid peroxidation in ferroptosis remain uncertain.
The term "ferroptosis" refers to a type of programmed cell death System Xc− , composed of SLC7A11 and SLC3A2, exchanges gluta­
brought on by perturbations of the intracellular microenvironment, mate for cystine in a 1:1 ratio in a normal state to enable the release of
especially the accumulation of toxic lipid peroxidation, which depends glutamate into the extracellular environment and conversion of cystine
on ROS production and iron availability [148]. Morphological changes into cysteine, which is the primary limiting substrate for the synthesis of
of ferroptotic cells include a distinctive rounded shape before cell death, glutathione [150]. As a reductant, glutathione peroxidase 4 (GPX4)
a complete nucleus without condensation, and smaller mitochondria mediates the reduction of phospholipid hydroperoxides (PLOOHs)
with increased membrane density [149]. Ferroptosis may be brought on [153]. GPX4 converts lipid hydroperoxides to lipid alcohols, thereby
by the buildup of glutamate, iron, and polyunsaturated fatty acids avoiding the iron (Fe2+)-dependent generation of harmful lipid ROS.
(PUFAs) or by the depletion of glutathione (GSH) and nicotinamide Consequently, when GPX4 is inhibited, PLOOHs persist longer and
adenine dinucleotide phosphate (NADPH) [150]. Instead, iron chelators, interact with both Fe2+ and Fe3+ to produce free radicals such as PLO•
lipid peroxidation inhibitors, and a reduction in polyunsaturated fatty and PLOO•, fueling the damaging peroxidation chain reaction (Fig. 6)
acids can all inhibit ferroptosis [151]. [154].
The combination of iron-bound transferrin and transferrin receptor Apart from the cysteine-GSH-GPX4 axis, recent studies have unveiled
(TFR) induces the import of Fe3+, which is subsequently reduced to GPX4-independent mechanisms of ferroptosis, including the NAD(P)H-
Fe2+ and transported into the cytosol via divalent metal transporter 1 ferroptosis suppressor protein 1-ubiquinone (NAD(P)H-FSP1-CoQ10)

Fig. 6. Ferroptosis signaling pathway in pregnancy related diseases.The ferroptosis signaling pathway is characterized by iron overload, lipid peroxidation, and
impaired GPX4 activity. Upon transportation into cells by TFR1 and DMT1 and subsequent reduction by STEAP3, excess iron participates in the production of ROS,
thereby promoting lipid peroxidation. Dysfunctional ferritin light chain, as observed in PE, aggravate the buildup of intracellular iron and oxidative stress. In
pregnancy related diseases, dysfunction of system Xc− and depletion of GSH lead to the inhibition of GPX4 activity, thus failing to counteract lipid peroxidation,
resulting in structural and functional impairment of trophoblast cells. Cellular metabolic disorders caused by excess sugar and mitochondrial stress also boost the
process of lipid peroxidation.(STEAP3, six-transmembrane epithelial antigen of prostate 3; FTL, ferritin light chain; GLUT, glucose transporter; Glu, glutamicacid).

10
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

pathway [155]. These findings highlight the complexity of ferroptosis fetal demise [171]. Both low serum iron levels and decidual local iron
regulation and suggest that more ferroptosis signaling pathways with deposition have been observed in patients with recurrent pregnancy loss
distinct biological functions are awaiting exploration. (RPL). Iron death inhibitors, cysteine, and GSH supplementation have
shown promise in lowering decidual stromal cell ferroptosis and
6.2. Ferroptosis in pregnancy related diseases reversing embryonic loss in spontaneous abortion models [172].
Therefore, ferroptosis in decidual stromal cells may be involved in the
6.2.1. Ferroptosis in PE pathogenesis of RPL. According to the research by Fang et al., GPX4 and
Based on the information provided, it’s evident that iron overload the expression of epidermal growth factor receptor, which has been
and ROS are closely associated with ferroptosis. A cross-sectional study demonstrated to participate in the regulation of ferroptosis, are both
revealed significantly elevated serum levels of iron, ferritin, transferrin elevated in the ICP group, suggesting the relationship between ferrop­
saturation, hepcidin, and lipid peroxidation byproducts like malon­ tosis and ICP [173].
dialdehyde in PE patients [156]. Reduced circulation selenium, GPX4
activity, and lower placental GSH levels in PE suggest that insufficient 7. Cell death-based therapeutic implications
antioxidant defenses may cause oxidative stress and the development of
PE [157,158]. The sudden increase in oxygen and iron resulting from Cell death is essential for morphogenesis and homeostasis mainte­
hypoxia/reperfusion events between the 8th and 10th weeks of preg­ nance, as it facilitates the removal of contaminated cells to prevent
nancy causes membrane lipid peroxidation and overactivated ferropto­ pathogens from proliferating. Impaired or over-activated cell death
sis at the maternal-fetal interface. This phenomenon contributes to pathways contribute to the occurrence of pregnancy related diseases.
shallow endovascular EVCT infiltration and defective uterine spiral ar­ Therefore, therapeutics for treating related diseases by correcting cell
tery remodeling, which are pathologic characteristics of PE [159]. death imbalances are burgeoning (Table 1).
Recent studies have shown that FTL increases intracellular iron accu­ Evidence indicates that moderate maternal nutritional intake during
mulation, promoting oxidative stress-induced ferroptosis and defective pregnancy can prevent illnesses by inhibiting apoptosis. Hyper­
uterine spiral artery remodeling [160]. Furthermore, the promotion of homocysteinemia, characterized by increased oxidative stress, can
ferroptosis and oxidative stress in PE may be mediated by an interaction trigger apoptosis. However, early supplementation with folate, vitamin
between elevated expression of the transmembrane channel protein B12, and long-chain polyunsaturated fatty acids can prevent the devel­
pannexin 1 and activated TLR4 [161]. Evidence suggests that opment of hyperhomocysteinemia, thereby reducing the risk of pre­
miR-30b-5p overexpression in PE is essential for ferroptosis. Concretely, eclampsia [38]. According to research, saturated free fatty acids
the upregulation of miR-30b-5p leads to the downregulation of SLC7A11 promote placental trophoblast apoptosis through activating caspase-3
and paired box 3 expression, further causing decreased levels of ferro­ and -7, which can be counteracted by monounsaturated fatty acids,
portin 1 and intracellular GSH levels, which in turn leads to the buildup indicating the therapeutic potential of monounsaturated fatty acids
of free iron within cells. The decreased glutathione and iron accumu­ [174]. Studies have also found that short-chain fatty acids promote
lation then trigger ferroptosis, ultimately compromising the function of macrophage autophagy, thus inhibiting macrophage inflammation and
the trophoblast [162]. alleviating PE [175]. Moreover, an in vitro study has found that vitamin
D can increase the viability of HUVEC, which suggests that vitamin D
6.2.2. Ferroptosis in GDM may be propitious for decreasing inflammation and cell death in the
The correlation between diabetes and iron overload significantly placenta of PE patients [176].
influences disease progression. Excessive iron levels damage the Some hormones and medications are believed to alleviate pregnancy
pancreatic β-cells through oxidative stress, leading to insulin resistance related diseases by restoring the balance of cell death. Progesterone
and elevating the susceptibility to diabetes; meanwhile, diabetes pro­ stimulates trophoblast invasion and suppresses apoptosis, thereby alle­
motes iron accumulation, which raises ferritin levels [163,164]. In viating PE-like symptoms caused by SIRT1 deficiency [177]. Melatonin
GDM, hyperglycemia circumstances stimulate the upregulation of has been shown in studies to decrease HTRA1 transcription via the
SIRT3, which causes trophoblast ferroptosis by enhancing the miR-520c-3p/SETD7 axis, enhancing invasion and migration of
AMPK-mTOR pathway and suppressing cellular GPX4 levels [165]. trophoblast and inhibiting trophoblasts pyroptosis in PE [120]. Several
Abnormal lipid metabolism frequently coexists with aberrant glucose natural compounds, such as atractylenolide and puerarin, attenuate
metabolism in patients with diabetes mellitus [166]. High levels of free apoptosis and oxidative stress in PE by activating or inhibiting different
fatty acids in GDM patients hinder insulin-induced glucose uptake, signaling pathways [178,179]. Ferulic acid, derived from plants, may be
which further strengthens insulin resistance and increases the risk of capable of treating PE through preventing cell damage and apoptosis
GDM [167]. Adiponectin, an insulin sensitizer that decreases in GDM induced by oxidative stress and inflammation [32]. According to recent
patients, weakens the effects of fatty acid oxidation/peroxide reports, autophagy maintenance may be a possible treatment target for
imbalance-mediated ferroptosis and ameliorates placental damage PE. Cyclosporine A, an immunosuppressant, may boost EVT prolifera­
caused by hyperlipidemia and hyperglycemia [164]. Additionally, tion, migration, and invasion by triggering autophagy while also pro­
recent research demonstrates that overexpressed CircHIPK3 in GDM tecting trophoblasts from oxidative stress [180]. However, excessive
patients promotes ferroptosis through miR-1278/DNMT1 to control autophagy caused by long-term hypoxia may induce cell death and
GPX4 DNA methylation [168]. impair placental function. Esomeprazole relieves PE-like symptoms by
blocking excessive placental autophagy through the SIRT1/AMPKα-m­
6.2.3. Ferroptosis in other pregnancy related diseases TOR pathway, preventing the release of toxic substances detrimental to
The role of ferroptosis in other pregnancy related diseases also re­ the maternal vasculature [84]. Excess iron and the buildup of fatal lipid
ceives significant interest. High hemoglobin concentrations caused by molecules from lipid peroxidation play an indispensable part in fer­
excess iron intake or high iron levels during pregnancy may increase roptosis. Iron chelators (e.g., deferiprone, deferoxamine), small lipo­
blood viscosity and damage placental blood flow, thus increasing the philic antioxidants (e.g., ferritetin, liplastin), and GSH and cystine (the
risk of preterm birth [169]. Uterine contractions, a defining feature of raw material for GSH production) can all contribute to reducing the risk
spontaneous preterm birth, are associated with placental hypoxia/r­ of ferroptosis-induced illnesses and improving pregnancy outcomes
eoxygenation and an increase in lipid mediators and related enzymes, [149,172].
both of which are risk factors for ferroptosis [170]. Furthermore, Furthermore, a range of signaling pathways, protein molecules,
Decreased GPX4 expression in placental trophoblast cells in spontaneous genes, and RNAs involved in cell death represent potential targets for the
abortion patients suggests a potential role of ferroptosis in first-trimester therapy of pregnancy related diseases. IL-15, an inflammatory factor

11
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

Table 1
Cell death-based therapeutic implications for pregnancy related diseases.
Cell death Drugs or compounds Mechanisms Diseases Refs

Apoptosis folate, vitamin B12, and long chain Preventing hyperhomocysteinemia PE [38]
polyunsaturated fatty acids
monounsaturated fatty acids Inhibiting activation of caspase-3 and -7 PE, GDM, IUGR and maternal [174]
inflammation
vitamin D Decreasing inflammation and cell death PE [176]
progesterone Stimulating trophoblast invasion and suppresses apoptosis PE [177]
atractylenolide Inhibiting apoptosis and oxidative stress by activating MAPK/ PE [178]
ERK signalling
puerarin Alleviating inflammation and apoptosis PE [179]
ferulic acid Preventing cell damage and apoptosis PE [32]
CLEC4GP1 Inhibits IL-15 mRNA stability PE [181]
ERp29 suppression Inhibiting phosphorylation and kinase activity of p38, thus ICP [183]
reversing the harmful effects of taurocholic acid
acetylcholine Inhibiting activation of p38 MAPK and NF-κB PE [184]
Autophagy short-chain fatty acids Promoting macrophage autophagy PE [175]
cyclosporine A Boosting EVT proliferation, migration, and invasion by PE [180]
triggering autophagy
esomeprazole Blocking excessive placental autophagy through the SIRT1/ PE [84]
AMPKα-mTOR pathway
Pyroptosis melatonin Decreasing HTRA1 transcription via the miR-520c-3p/SETD7 PE [120]
axis
metformin Blocking TLR4/NF-κB signaling PE [123]
kisspeptin-10 Prevents the activation of the inflammasome-NLRP3 and the gestational diseases that involve [125]
placental pyroptosis inflammasome activation and pyroptosis
Ferroptosis deferiprone, deferoxamine, ferritetin, Inhibiting ferroptosis PE, GDM, RPL and spontaneous preterm [149,
liplastin, GSH and cystine birth 172]

found to be upregulated in PE placenta, exerts detrimental effects. A intolerance, which increases the risk of diabetes and other near- and
pseudogene named C-type lectin domain family 4 member G pseudo­ long-term complications in pregnant women and fetuses. Meanwhile,
gene 1 inhibits IL-15 mRNA stability, which in turn inhibits trophoblast pregnancy related diseases can exacerbate the dysregulation of cell
cell apoptosis and promotes invasion [181]. p38 MAPKs, death, creating a vicious cycle that ultimately results in adverse preg­
serine/threonine-specific protein kinases, allow cells to react to stimuli nancy outcomes. Therefore, studies in mounting numbers have put great
and influence cell proliferation, apoptosis, autophagy, and tumor emphasis on the role of cell death in pregnancy related diseases, espe­
growth [182]. According to research, ERp29 suppression could reduce cially the newly discovered pyroptosis, ferroptosis, and necroptosis, in
apoptosis in ICP through inhibiting the phosphorylation and kinase ac­ order to provide potential therapeutic strategies targeting relevant
tivity of p38, therefore reversing the harmful effects of taurocholic acid signaling pathways.
[183]. Acetylcholine may weaken hypoxia-induced oxidative stress and However, the mechanisms of cell death have yet to be fully eluci­
apoptosis via inhibiting activation of p38 MAPK and NF-κB, suggesting dated, and the signaling pathways involved in the development of
that increased vagal activity may be beneficial for treating PE [184]. The pregnancy related diseases remain unclear. Among them, there are
activation of NLRP3 inflammasomes serves as a vital link in pyroptosis, fewer studies on the relationship between necroptosis and pregnancy
which occurs in pregnancy illnesses such as PE and GDM. Thus, NLRP3 related disorders, resulting in a lack of understanding in this area. In the
inhibitors may be an alternative for treatment [119]. For example, meantime, most of the current treatment strategies for pregnancy
through blocking TLR4/NF-κB signaling in a PE trophoblast model, related diseases based on cell death are derived from in vitro experi­
metformin inhibits the pyroptosis triggered by the NLRP3 inflamma­ ments and have not yet entered clinical trials. Consequently, it is still
some [123]. A recent study has found that kisspeptin-10 hinders the unknown whether these strategies can be effectively applied in clinical
activation of the NLRP3 inflammasome and placental pyroptosis practice. We hope that deeper studies in the future will reveal the po­
brought on by maternal hypothyroidism, implying that kisspeptin ana­ tential mechanisms linking cell death and pregnancy related diseases
logues may hold promise for managing pregnancy diseases associated and provide valuable insights for the development of meaningful ther­
with pyroptosis [125]. apeutic strategies.
More regulators and effectors of cell death pathways open up new
avenues for therapeutic intervention and are expected to be applied in Ethics approval and consent to participate
clinical practice, contingent on further research on the underlying
mechanisms of pregnancy related diseases. Not applicable.

8. Conclusions and future perspectives Consent for publication

Cell death is indispensable for the regular growth of the organism as Not applicable.
well as maternal health and fetal development throughout pregnancy, as
it allows for the removal of extra or damaged cells produced by high
CRediT authorship contribution statement
proliferation along with the optimal formation and maturation of tissues
and organs through the elimination of undesired cells. Nevertheless,
Yan Zhang, Ru Zhu and Yun Zhang conceived this project. Xiaowen
abnormalities in trophoblast cell death lead to a homeostatic imbalance
Xie, Jiayu Liu, Jingyi Gao and Chenwei Shang wrote the original draft.
of local placental tissue, disrupting trophoblast cell migration and in­
Xiaowen Xie and Chenwei Shang designed the figures. Chenwei Shang,
vasion and spiral artery remodeling, consequently resulting in preg­
Ying Jiang, Lingyan Chen, Zhiwen Qian, Lu Liu and Danping Wu
nancy related diseases. Excessive cell death of maternal pancreatic β
collected the data and performed the analysis. Yan Zhang and Xiaowen
cells leads to impaired pancreatic β cell function and maternal glucose
Xie revised the manuscript. Yan Zhang got financial support. All authors

12
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

reviewed and approved the final manuscript. [22] P.J. Jost, D. Vucic, Regulation of cell death and immunity by XIAP, Cold Spring
Harb. Perspect. Biol. 12 (2020) a036426.
[23] Q.-H. Yang, R. Church-Hajduk, J. Ren, M.L. Newton, C. Du, Omi/HtrA2 catalytic
Funding cleavage of inhibitor of apoptosis (IAP) irreversibly inactivates IAPs and
facilitates caspase activity in apoptosis, Genes Dev. 17 (2003) 1487–1496.
[24] D. Bertheloot, E. Latz, B.S. Franklin, Necroptosis, pyroptosis and apoptosis: an
The study was partially supported by Major project of Wuxi Science intricate game of cell death, Cell. Mol. Immunol. 18 (2021) 1106–1121.
and Technology Bureau (N20201006), Wuxi Double-Hundred Talent [25] K.M. Boatright, G.S. Salvesen, Mechanisms of caspase activation, Curr. Opin. Cell
Fund Project (BJ2023075), Wuxi Health Commission Precision Medi­ Biol. 15 (2003) 725–731.
[26] N. Ozören, W.S. El-Deiry, Defining characteristics of Types I and II apoptotic cells
cine Project (J202106), Jiangsu Provincial Maternal and Child Health in response to TRAIL, Neoplasia N. Y. N. 4 (2002) 551–557.
Research Project (F202034) and Jiangsu Provincial Six Talent Peaks [27] M.-X. He, Y.-W. He, A role for c-FLIPL in the regulation of apoptosis, autophagy,
Project (No. YY-124). and necroptosis in T lymphocytes, Cell Death Differ. 20 (2013) 188–197.
[28] R. Iurlaro, C. Muñoz-Pinedo, Cell death induced by endoplasmic reticulum stress,
FEBS J. 283 (2016) 2640–2652.
[29] H. Hu, M. Tian, C. Ding, S. Yu, The C/EBP homologous protein (CHOP)
Declaration of Competing Interest transcription factor functions in endoplasmic reticulum stress-induced apoptosis
and microbial infection, Front. Immunol. 9 (2018) 3083.
The authors declare that they have no known competing financial [30] S. Rana, E. Lemoine, J.P. Granger, S.A. Karumanchi, Preeclampsia:
pathophysiology, challenges, and perspectives, Circ. Res. 124 (2019) 1094–1112.
interests or personal relationships that could have appeared to influence [31] E.A. Phipps, R. Thadhani, T. Benzing, S.A. Karumanchi, Pre-eclampsia:
the work reported in this paper. pathogenesis, novel diagnostics and therapies, Nat. Rev. Nephrol. 15 (2019)
275–289.
[32] Y. Chen, et al., Ferulic acid ameliorated placental inflammation and apoptosis in
Data availability rat with preeclampsia, Clin. Exp. Hypertens. N. Y. N. 1993 41 (2019) 524–530.
[33] H. Liu, W. Wang, C. Liu, Increased expression of IFN-γ in preeclampsia impairs
No data was used for the research described in the article. human trophoblast invasion via a SOCS1/JAK/STAT1 feedback loop. Exp. Ther.
Med. 21 (2021) 112.
[34] V. Kasture, A. Kale, K. Randhir, D. Sundrani, S. Joshi, Effect of maternal omega-3
Acknowledgements fatty acids and vitamin E supplementation on placental apoptotic markers in rat
model of early and late onset preeclampsia, Life Sci. 239 (2019) 117038.
[35] S.D. Smith, C.E. Dunk, J.D. Aplin, L.K. Harris, R.L. Jones, Evidence for immune
The figures in this manuscript were created using BioRender, a cell involvement in decidual spiral arteriole remodeling in early human
valuable tool for scientific illustrations. We would like to thank Bio­ pregnancy, Am. J. Pathol. 174 (2009) 1959–1971.
Render for granting us permission to use their images in this publication. [36] T. Cindrova-Davies, O. Spasic-Boskovic, E. Jauniaux, D.S. Charnock-Jones, G.
J. Burton, Nuclear factor-kappa B, p38, and stress-activated protein kinase
mitogen-activated protein kinase signaling pathways regulate proinflammatory
References cytokines and apoptosis in human placental explants in response to oxidative
stress: effects of antioxidant vitamins, Am. J. Pathol. 170 (2007) 1511–1520.
[37] P. Wang, et al., Resveratrol induces SIRT1-dependent autophagy to prevent
[1] M.S. D’Arcy, Cell death: a review of the major forms of apoptosis, necrosis and
H2O2-Induced oxidative stress and apoptosis in HTR8/SVneo cells, Placenta 91
autophagy, Cell Biol. Int. 43 (2019) 582–592.
(2020) 11–18.
[2] I.K.H. Poon, C.D. Lucas, A.G. Rossi, K.S. Ravichandran, Apoptotic cell clearance:
[38] V.V. Kasture, D.P. Sundrani, S.R. Joshi, Maternal one carbon metabolism through
basic biology and therapeutic potential, Nat. Rev. Immunol. 14 (2014) 166–180.
increased oxidative stress and disturbed angiogenesis can influence placental
[3] Mechanism and disease implications of necroptosis and neuronal inflammation -
apoptosis in preeclampsia, Life Sci. 206 (2018) 61–69.
PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/30185777/〉.
[39] L. Teirilä, J. Heikkinen-Eloranta, J. Kotimaa, S. Meri, A.I. Lokki, Regulation of the
[4] M. Fricker, A.M. Tolkovsky, V. Borutaite, M. Coleman, G.C. Brown, Neuronal Cell
complement system and immunological tolerance in pregnancy, Semin. Immunol.
Death, Physiol. Rev. 98 (2018) 813–880.
45 (2019) 101337.
[5] FAS and RAS related Apoptosis defects: From autoimmunity to leukemia -
[40] S. Liu, S. Jiang, L. Huang, Y. Yu, Expression of SASH1 in preeclampsia and its
Meynier - 2019 - Immunological Reviews - Wiley Online Library. 〈https://onlineli
effects on human trophoblast, BioMed. Res. Int. 2020 (2020) 5058260.
brary.wiley.com/doi/10.1111/imr.12720〉.
[41] J. Munjas, et al., Non-coding RNAs in preeclampsia-molecular mechanisms and
[6] Cell Death in the Origin and Treatment of Cancer - PubMed. 〈https://pubmed.
diagnostic potential, Int. J. Mol. Sci. 22 (2021) 10652.
ncbi.nlm.nih.gov/32516599/〉.
[42] H. Zheng, et al., Dysregulated pseudogene BNIP3P1 inhibited cell proliferation
[7] Regulation of Placental Extravillous Trophoblasts by the Maternal Uterine
and promoted cell apoptosis in preeclampsia by acting as a competing
Environment - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/30483261/〉.
endogenous RNA for BNIP3, Environ. Toxicol. 37 (2022) 971–982.
[8] V. Kasture, A. Sahay, S. Joshi, Cell death mechanisms and their roles in pregnancy
[43] K.R. Palmer, L. Xiaohua, B.W. Mol, Management of intrahepatic cholestasis in
related disorders, Adv. Protein Chem. Struct. Biol. 126 (2021) 195–225.
pregnancy, Lancet Lond. Engl. 393 (2019) 853–854.
[9] D. Shan, R. Dong, Y. Hu, Current understanding of autophagy in intrahepatic
[44] T. Zhang, et al., Comparative proteomics analysis of placenta from pregnant
cholestasis of pregnancy, Placenta 115 (2021) 53–59.
women with intrahepatic cholestasis of pregnancy, PloS One 8 (2013) e83281.
[10] Reduced apoptosis in term placentas from gestational diabetic pregnancies -
[45] MicroRNA-Mediated Control of Inflammation and Tolerance in Pregnancy -
PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/25054844/〉.
PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/31024550/〉.
[11] T.R. Magee, et al., Gestational diabetes mellitus alters apoptotic and
[46] Serum Exosomes MicroRNAs Are Novel Non-Invasive Biomarkers of Intrahepatic
inflammatory gene expression of trophobasts from human term placenta,
Cholestasis of Pregnancy - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/356005
J. Diabetes Complicat. 28 (2014) 448–459.
87/〉.
[12] A.W. Lokeswara, R. Hiksas, R. Irwinda, N. Wibowo, Preeclampsia: from cellular
[47] L.X. Ji, J. Liu, [MicroRNA-221/222 participates in the pathogenesis of
wellness to inappropriate cell death, and the roles of nutrition, Front. Cell Dev.
intrahepatic cholestasis of pregnancy via promoting the apoptosis of human
Biol. 9 (2021) 726513.
placental trophoblast HTR-8 cells], Zhonghua Gan Zang Bing. Za Zhi Zhonghua
[13] O. Beharier, K. Kajiwara, Y. Sadovsky, Ferroptosis, trophoblast lipotoxic damage,
Ganzangbing Zazhi Chin. J. Hepatol. 26 (2018) 607–611.
and adverse pregnancy outcome, Placenta 108 (2021) 32–38.
[48] F. Feng, L. Lei, J. Liao, X. Huang, Y. Shao, Circ_0060731 mediated miR-21-5p-
[14] J.F.R. Kerr, A.H. Wyllie, A.R. Currie, Apoptosis: a basic biological phenomenon
PDCD4/ESR1 pathway to induce apoptosis of placental trophoblasts in
with wideranging implications in tissue kinetics, Br. J. Cancer 26 (1972)
intrahepatic cholestasis of pregnancy, Tissue Cell 76 (2022) 101771.
239–257.
[49] M. Brecker, S. Khakhina, T.J. Schubert, Z. Thompson, R.C. Rubenstein, The
[15] N. Ketelut-Carneiro, K.A. Fitzgerald, Apoptosis, pyroptosis, and necroptosis-oh
probable, possible, and novel functions of ERp29, Front. Physiol. 11 (2020)
my! the many ways a cell can die, J. Mol. Biol. 434 (2022) 167378.
574339.
[16] J.G. Nirmala, M. Lopus, Cell death mechanisms in eukaryotes, Cell Biol. Toxicol.
[50] T. Zhang, et al., High concentraction of taurocholic acid induced apoptosis in
36 (2020) 145–164.
HTR-8/SVneo cells via overexpression of ERp29 and activation of p38, Placenta
[17] F.J. Bock, S.W.G. Tait, Mitochondria as multifaceted regulators of cell death, Nat.
35 (2014) 496–500.
Rev. Mol. Cell Biol. 21 (2020) 85–100.
[51] H.-Z. Wang, et al., [Role of endoplasmic reticulum stress-induced apoptosis of
[18] J. Prudent, et al., MAPL SUMOylation of Drp1 Stabilizes an ER/Mitochondrial
trophoblasts in intrahepatic cholestasis during pregnancy, Nan Fang. Yi Ke Da
Platform Required for Cell Death, Mol. Cell 59 (2015) 941–955.
Xue Xue Bao 38 (2018) 572–577.
[19] K. Huang, et al., BH3-only proteins target BCL-xL/MCL-1, not BAX/BAK, to
[52] Z. Song, X. Tian, Q. Shi, Fas, Caspase-8, and Caspase-9 pathway-mediated bile
initiate apoptosis, Cell Res 29 (2019) 942–952.
acid-induced fetal cardiomyocyte apoptosis in intrahepatic cholestasis pregnant
[20] S.B. Bratton, et al., Recruitment, activation and retention of caspases-9 and -3 by
rat models, J. Obstet. Gynaecol. Res. 47 (2021) 2298–2306.
Apaf-1 apoptosome and associated XIAP complexes, EMBO J. 20 (2001)
[53] A. Sweeting, J. Wong, H.R. Murphy, G.P. Ross, A clinical update on gestational
998–1009.
diabetes mellitus, Endocr. Rev. 43 (2022) 763–793.
[21] D. Acehan, et al., Three-dimensional structure of the apoptosome: implications
for assembly, procaspase-9 binding, and activation, Mol. Cell 9 (2002) 423–432.

13
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

[54] D. Shen, et al., Mechanism of neutrophil extracellular traps generation and their [89] L. Gao, et al., Glycochenodeoxycholate promotes hepatocellular carcinoma
role in trophoblasts apoptosis in gestational diabetes mellitus, Cell. Signal. 88 invasion and migration by AMPK/mTOR dependent autophagy activation, Cancer
(2021) 110168. Lett. 454 (2019) 215–223.
[55] X. Liao, Z. Zhou, X. Zhang, Effects of miR‑195‑5p on cell proliferation and [90] Glycochenodeoxycholic acid impairs transcription factor E3 -dependent
apoptosis in gestational diabetes mellitus via targeting EZH2, Mol. Med. Rep. 22 autophagy-lysosome machinery by disrupting reactive oxygen species
(2020) 803–809. homeostasis in L02 cells - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/32
[56] Y.-L. Zhang, X.-Q. Chen, Dysregulation of microRNA-770-5p influences 439580/〉.
pancreatic-β-cell function by targeting TP53 regulated inhibitor of apoptosis 1 in [91] K. Panzitt, P. Fickert, M. Wagner, Regulation of autophagy by bile acids and in
gestational diabetes mellitus, Eur. Rev. Med. Pharmacol. Sci. 24 (2020) 793–801. cholestasis - CholestoPHAGY or CholeSTOPagy, Biochim. Biophys. Acta Mol.
[57] J. Wen, X. Bai, miR-520h Inhibits cell survival by targeting mTOR in gestational Basis Dis. 1867 (2021) 166017.
diabetes mellitus, Acta Biochim. Pol. 68 (2021) 65–70. [92] J.F. Plows, J.L. Stanley, P.N. Baker, C.M. Reynolds, M.H. Vickers, The
[58] Pregnancy-associated cardiac dysfunction and the regulatory role of microRNAs - pathophysiology of gestational diabetes mellitus, Int. J. Mol. Sci. 19 (2018) 3342.
PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/32252821/〉. [93] A.D. Barlow, D.C. Thomas, Autophagy in diabetes: β-cell dysfunction, insulin
[59] IL-34 causes inflammation and beta cell apoptosis and dysfunction in gestational resistance, and complications, DNA Cell Biol. 34 (2015) 252–260.
diabetes mellitus - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/31648183/〉. [94] L. Ji, et al., Systematic characterization of autophagy in gestational diabetes
[60] D.P. Damron, Definition of fetal growth restriction, Am. J. Obstet. Gynecol. 224 mellitus, Endocrinology 158 (2017) 2522–2532.
(2021) 242. [95] X. Bai, et al., CAV1-CAVIN1-LC3B-mediated autophagy regulates high glucose-
[61] Y. Lv, et al., Down-regulated expressed protein HMGB3 inhibits proliferation and stimulated LDL transcytosis, Autophagy 16 (2020) 1111–1129.
migration, promotes apoptosis in the placentas of fetal growth restriction, Int. J. [96] Y. Bao, J. Zhang, Y. Liu, L. Wu, J. Yang, Identification of human placenta-derived
Biochem. Cell Biol. 107 (2019) 69–76. circular RNAs and autophagy related circRNA-miRNA-mRNA regulatory network
[62] H.-L. Zhu, et al., Gestational exposure to environmental cadmium induces in gestational diabetes mellitus, Front. Genet. 13 (2022) 1050906.
placental apoptosis and fetal growth restriction via Parkin-modulated MCL-1 [97] L. Hong, et al., Multi-omics reveals a relationship between endometrial amino
degradation, J. Hazard. Mater. 424 (2022) 127268. acid metabolism and autophagy in women with recurrent miscarriage†, Biol.
[63] J. Cui, et al., miR-1227-3p participates in the development of fetal growth Reprod. 105 (2021) 393–402.
restriction via regulating trophoblast cell proliferation and apoptosis, Sci. Rep. 12 [98] H.-L. Zhu, et al., Melatonin protects against environmental stress-induced fetal
(2022) 6374. growth restriction via suppressing ROS-mediated GCN2/ATF4/BNIP3-dependent
[64] P. Kovács, et al., The role of apoptosis in the complex pathogenesis of the most mitophagy in placental trophoblasts, Redox Biol. 40 (2021) 101854.
common obstetrics and gynaecology diseases, Physiol. Int. 107 (2020) 106–119. [99] X. Zhao, et al., Physiological and pathological regulation of autophagy in
[65] D. Sun, H. Wu, Z. Ping, H. Zhu, L. Ai, PLAC1 regulates the occurrence of fetal pregnancy, Arch. Gynecol. Obstet. 302 (2020) 293–303.
growth restriction by inhibiting the apoptosis of trophoblast cells, Ann. Clin. Lab. [100] Y. Fang, et al., Pyroptosis: a new frontier in cancer, Biomed. Pharmacother.
Sci. 51 (2021) 182–189. Biomedecine Pharmacother. 121 (2020) 109595.
[66] S. Quenby, et al., Miscarriage matters: the epidemiological, physical, [101] X. Xia, et al., The role of pyroptosis in cancer: pro-cancer or pro-"host", Cell Death
psychological, and economic costs of early pregnancy loss, Lancet Lond. Engl. 397 Dis. 10 (2019) 650.
(2021) 1658–1667. [102] D. Frank, J.E. Vince, Pyroptosis versus necroptosis: similarities, differences, and
[67] S. Qin, et al., SPRY4 regulates trophoblast proliferation and apoptosis via crosstalk, Cell Death Differ. 26 (2019) 99–114.
regulating IFN-γ-induced STAT1 expression and activation in recurrent [103] Y. He, H. Hara, G. Núñez, Mechanism and regulation of NLRP3 inflammasome
miscarriage. Am. J. Reprod. Immunol. N. Y. N. 1989 83 (2020) e13234. activation, Trends Biochem. Sci. 41 (2016) 1012–1021.
[68] X. Li, et al., Downregulation of CCNA2 disturbs trophoblast migration, [104] H.T.H. Thi, S. Hong, Inflammasome as a therapeutic target for cancer prevention
proliferation, and apoptosis during the pathogenesis of recurrent miscarriage, and treatment, J. Cancer Prev. 22 (2017) 62–73.
Am. J. Reprod. Immunol. N. Y. N. 1989 82 (2019) e13144. [105] L. A, M. Sl, Homeostasis-altering molecular processes as mechanisms of
[69] Monocytes and macrophages in pregnancy: The good, the bad, and the ugly - inflammasome activation, Nat. Rev. Immunol. 17 (2017).
PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/35451089/〉. [106] A. Zahid, B. Li, A.J.K. Kombe, T. Jin, J. Tao, Pharmacological Inhibitors of the
[70] J. Ding, T. Yin, N. Yan, Y. Cheng, J. Yang, FasL on decidual macrophages NLRP3 Inflammasome, Front. Immunol. 10 (2019) 2538.
mediates trophoblast apoptosis: a potential cause of recurrent miscarriage, Int. J. [107] G. Sollberger, G.E. Strittmatter, M. Garstkiewicz, J. Sand, H.-D. Beer, Caspase-1:
Mol. Med. 43 (2019) 2376–2386. the inflammasome and beyond, Innate Immun. 20 (2014) 115–125.
[71] Y. Yao, F. Hao, L.-C. Tang, X.-H. Xu, L. Jin, Downregulation of HDAC8 expression [108] L. Sborgi, et al., GSDMD membrane pore formation constitutes the mechanism of
decreases CD163 levels and promotes the apoptosis of macrophages by activating pyroptotic cell death, EMBO J. 35 (2016) 1766–1778.
the ERK signaling pathway in recurrent spontaneous miscarriage, Mol. Hum. [109] X. Chen, et al., Pyroptosis is driven by non-selective gasdermin-D pore and its
Reprod. 26 (2020) 521–531. morphology is different from MLKL channel-mediated necroptosis, Cell Res 26
[72] Autophagy: cellular and molecular mechanisms - PubMed. 〈https://pubmed.ncbi. (2016) 1007–1020.
nlm.nih.gov/20225336/〉. [110] X. Liu, et al., Inflammasome-activated gasdermin D causes pyroptosis by forming
[73] D. Denton, S. Kumar, Autophagy-dependent cell death, Cell Death Differ. 26 membrane pores, Nature 535 (2016) 153–158.
(2019) 605–616. [111] M. Jiang, L. Qi, L. Li, Y. Li, The caspase-3/GSDME signal pathway as a switch
[74] D.R. Green, F. Llambi, Cell death signaling, Cold Spring Harb. Perspect. Biol. 7 between apoptosis and pyroptosis in cancer, Cell Death Discov. 6 (2020) 112.
(2015) a006080. [112] L.H. Chu, et al., The oxidized phospholipid oxPAPC protects from septic shock by
[75] Y.C. Kim, K.-L. Guan, mTOR: a pharmacologic target for autophagy regulation, targeting the non-canonical inflammasome in macrophages, Nat. Commun. 9
J. Clin. Invest. 125 (2015) 25–32. (2018) 996.
[76] S. Alers, A.S. Löffler, S. Wesselborg, B. Stork, Role of AMPK-mTOR-Ulk1/2 in the [113] J. Shi, W. Gao, F. Shao, Pyroptosis: gasdermin-mediated programmed necrotic
regulation of autophagy: cross talk, shortcuts, and feedbacks, Mol. Cell. Biol. 32 cell death, Trends Biochem. Sci. 42 (2017) 245–254.
(2012) 2–11. [114] S.B. Kovacs, E.A. Miao, Gasdermins: effectors of pyroptosis, Trends Cell Biol. 27
[77] Y. Ohashi, Activation mechanisms of the VPS34 complexes, Cells 10 (2021) 3124. (2017) 673–684.
[78] E. Dimitriadis, et al., Pre-eclampsia, Nat. Rev. Dis. Prim. 9 (2023) 8. [115] Pyroptosis and Apoptosis Pathways Engage in Bidirectional Crosstalk in
[79] A. Nakashima, et al., Impaired autophagy by soluble endoglin, under Monocytes and Macrophages - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/28
physiological hypoxia in early pregnant period, is involved in poor placentation 392147/〉.
in preeclampsia, Autophagy 9 (2013) 303–316. [116] J. Hou, et al., PD-L1-mediated gasdermin C expression switches apoptosis to
[80] X. Yue, et al., GRIM-19 inhibition induced autophagy through activation of ERK pyroptosis in cancer cells and facilitates tumour necrosis, Nat. Cell Biol. 22 (2020)
and HIF-1α not STAT3 in Hela cells, Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 1264–1275.
37 (2016) 9789–9796. [117] I. C Weel, et al., Increased expression of NLRP3 inflammasome in placentas from
[81] A. Farina, et al., Gene expression in chorionic villous samples at 11 weeks’ pregnant women with severe preeclampsia, J. Reprod. Immunol. 123 (2017)
gestation from women destined to develop preeclampsia. Prenat. Diagn. 28 40–47.
(2008) 956–961. [118] M.L. Matias, et al., Endogenous and uric acid-induced activation of NLRP3
[82] H. Zhang, et al., Autophagy attenuates placental apoptosis, oxidative stress and inflammasome in pregnant women with preeclampsia, PloS One 10 (2015)
fetal growth restriction in pregnant ewes, Environ. Int. 173 (2023) 107806. e0129095.
[83] SIRT1: A Novel Protective Molecule in Pre-eclampsia - PubMed. 〈https://pubmed. [119] P.R. Nunes, S.V. Mattioli, V.C. Sandrim, NLRP3 activation and its relationship to
ncbi.nlm.nih.gov/35813294/〉. endothelial dysfunction and oxidative stress: implications for preeclampsia and
[84] S. Gu, et al., Esomeprazole inhibits hypoxia/endothelial dysfunction-induced pharmacological interventions, Cells 10 (2021) 2828.
autophagy in preeclampsia, Cell Tissue Res 388 (2022) 181–194. [120] Z. Liu, B. Chen, J. Chang, L. Feng, X. Zhao, Melatonin regulates trophoblast
[85] A. Nakashima, et al., Autophagy regulation in preeclampsia: pros and cons, pyroptosis, invasion and migration in preeclampsia by inhibiting HtrA1
J. Reprod. Immunol. 123 (2017) 17–23. transcription through the microRNA-520c-3p/SETD7 axis, Am. J. Reprod.
[86] ANGPTL4 mediates the protective role of PPARγ activators in the pathogenesis of Immunol. N. Y. N. 1989 87 (2022) e13523.
preeclampsia - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/28933788/〉. [121] J. Liu, W. Yang, Mechanism of histone deacetylase HDAC2 in FOXO3-mediated
[87] B. Kong, et al., Mechanism of tissue-specific farnesoid X receptor in suppressing trophoblast pyroptosis in preeclampsia, Funct. Integr. Genom. 23 (2023) 152.
the expression of genes in bile-acid synthesis in mice, Hepatol. Baltim. Md 56 [122] S.-B. Cheng, et al., Pyroptosis is a critical inflammatory pathway in the placenta
(2012) 1034–1043. from early onset preeclampsia and in human trophoblasts exposed to hypoxia and
[88] K. Panzitt, et al., FXR-dependent Rubicon induction impairs autophagy in models endoplasmic reticulum stressors, Cell Death Dis. 10 (2019) 927.
of human cholestasis, J. Hepatol. 72 (2020) 1122–1131.

14
X. Xie et al. Biomedicine & Pharmacotherapy 176 (2024) 116815

[123] Y. Zhang, et al., Metformin corrects glucose metabolism reprogramming and [155] X. Tong, et al., Targeting cell death pathways for cancer therapy: recent
NLRP3 inflammasome-induced pyroptosis via inhibiting the TLR4/NF-κB/ developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research,
PFKFB3 signaling in trophoblasts: implication for a potential therapy of J. Hematol. Oncol. J. Hematol. Oncol. 15 (2022) 174.
preeclampsia. Oxid. Med. Cell. Longev. 2021 (2021) 1806344. [156] N. Shaji Geetha, Z. Bobby, G. Dorairajan, S.E. Jacob, Increased hepcidin levels in
[124] S.-Y. Yu, X.-L. Li, Pyroptosis and inflammasomes in obstetrical and gynecological preeclampsia: a protective mechanism against iron overload mediated oxidative
diseases, Gynecol. Endocrinol. Off. J. Int. Soc. Gynecol. Endocrinol. 37 (2021) stress, J. Matern. Fetal Neonatal Med. 35 (2022) 636–641.
385–391. [157] M. Hd, W. V, R. Mm, S. Me, B.P. F, Reduced selenium concentrations and
[125] B.R. Santos, et al., Kisspeptin suppresses inflammasome-NLRP3 activation and glutathione peroxidase activity in preeclamptic pregnancies, Hypertens. Dallas
pyroptosis caused by hypothyroidism at the maternal-fetal interface of rats, Int. J. Tex. 1979 52 (2008).
Mol. Sci. 24 (2023) 6820. [158] Y. Zhang, Y. Lu, L. Jin, Iron Metabolism and Ferroptosis in Physiological and
[126] Z. Zhao, et al., Zika virus causes placental pyroptosis and associated adverse fetal Pathological Pregnancy, Int. J. Mol. Sci. 23 (2022) 9395.
outcomes by activating GSDME, eLife 11 (2022) e73792. [159] S.-W. Ng, S.G. Norwitz, E.R. Norwitz, The impact of iron overload and ferroptosis
[127] J. Guo, et al., TIGAR deficiency induces caspase-1-dependent trophoblasts on reproductive disorders in humans: implications for preeclampsia, Int. J. Mol.
pyroptosis through NLRP3-ASC inflammasome, Front. Immunol. 14 (2023) Sci. 20 (2019) 3283.
1114620. [160] X. Yang, et al., Ferritin light chain deficiency-induced ferroptosis is involved in
[128] E.H. Joo, et al., Effect of endogenic and exogenic oxidative stress triggers on preeclampsia pathophysiology by disturbing uterine spiral artery remodelling,
adverse pregnancy outcomes: preeclampsia, fetal growth restriction, gestational Redox Biol. 58 (2022) 102555.
diabetes mellitus and preterm birth, Int. J. Mol. Sci. 22 (2021) 10122. [161] S.R.A. El-Khalik, R.R. Ibrahim, M.T.A. Ghafar, D. Shatat, O.S. El-Deeb, Novel
[129] H. Zou, et al., Destruction in maternal-fetal interface of URSA patients via the insights into the SLC7A11-mediated ferroptosis signaling pathways in
increase of the HMGB1-RAGE/TLR2/TLR4-NF-κB signaling pathway. Life Sci. 250 preeclampsia patients: identifying pannexin 1 and toll-like receptor 4 as
(2020) 117543. innovative prospective diagnostic biomarkers, J. Assist. Reprod. Genet. 39 (2022)
[130] D. Zhu, et al., Inhibition of HMGB1 ameliorates the maternal-fetal interface 1115–1124.
destruction in unexplained recurrent spontaneous abortion by suppressing [162] H. Zhang, et al., miR-30-5p-mediated ferroptosis of trophoblasts is implicated in
pyroptosis activation, Front. Immunol. 12 (2021) 782792. the pathogenesis of preeclampsia, Redox Biol. 29 (2020) 101402.
[131] T. Zhang, Y. Wang, H. Inuzuka, W. Wei, Necroptosis pathways in tumorigenesis, [163] Oxidative stress impairs insulin internalization in endothelial cells in vitro -
Semin. Cancer Biol. 86 (2022) 32–40. PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/11380079/〉.
[132] W. Gao, X. Wang, Y. Zhou, X. Wang, Y. Yu, Autophagy, ferroptosis, pyroptosis, [164] Y. Zheng, Q. Hu, J. Wu, Adiponectin ameliorates placental injury in gestational
and necroptosis in tumor immunotherapy, Signal Transduct. Target. Ther. 7 diabetes mice by correcting fatty acid oxidation/peroxide imbalance-induced
(2022) 196. ferroptosis via restoration of CPT-1 activity, Endocrine 75 (2022) 781–793.
[133] J. Seo, et al., The roles of ubiquitination in extrinsic cell death pathways and its [165] D. Han, et al., SIRT3 deficiency is resistant to autophagy-dependent ferroptosis by
implications for therapeutics, Biochem. Pharmacol. 162 (2019) 21–40. inhibiting the AMPK/mTOR pathway and promoting GPX4 levels, J. Cell. Physiol.
[134] Necroptosis: A Novel Pathway in Neuroinflammation - PubMed. 〈https://pubmed. 235 (2020) 8839–8851.
ncbi.nlm.nih.gov/34322024/〉. [166] J.G. Jones, Hepatic glucose and lipid metabolism, Diabetologia 59 (2016)
[135] M. Zheng, T.-D. Kanneganti, The regulation of the ZBP1-NLRP3 inflammasome 1098–1103.
and its implications in pyroptosis, apoptosis, and necroptosis (PANoptosis), [167] Plasma Levels of Free Fatty Acids in Women with Gestational Diabetes and Its
Immunol. Rev. 297 (2020) 26–38. Intrinsic and Extrinsic Determinants: Systematic Review and Meta-Analysis -
[136] J. Seo, Y.W. Nam, S. Kim, D.-B. Oh, J. Song, Necroptosis molecular mechanisms: PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/31531374/〉.
Recent findings regarding novel necroptosis regulators, Exp. Mol. Med. 53 (2021) [168] J. Jiang, et al., Circular RNA HIPK3 facilitates ferroptosis in gestational diabetes
1007–1017. mellitus by regulating glutathione peroxidase 4 DNA methylation, J. Gene Med.
[137] Necroptosis in the Pathophysiology of Disease - PubMed. 〈https://pubmed.ncbi. (2023) e3526, https://doi.org/10.1002/jgm.3526.
nlm.nih.gov/31783008/〉. [169] K.G. Dewey, B.M. Oaks, U-shaped curve for risk associated with maternal
[138] D. Ma, et al., The development of necroptosis: what we can learn, Cell Stress hemoglobin, iron status, or iron supplementation, Am. J. Clin. Nutr. 106 (2017)
Chaperon-.-. 28 (2023) 969–987. 1694S–1702S.
[139] H. Yu, L. Chen, B. Du, Necroptosis in the pathophysiology of preeclampsia, Cell [170] PLA2G6 guards placental trophoblasts against ferroptotic injury - PubMed. 〈http
Cycle Georget. Tex. 22 (2023) 1713–1725. s://pubmed.ncbi.nlm.nih.gov/33087576/〉.
[140] W. Lu, H. Ji, D. Wu, SIRT2 plays complex roles in neuroinflammation [171] X. Shen, N. Obore, Y. Wang, T. Yu, H. Yu, The role of ferroptosis in placental-
neuroimmunology-associated disorders, Front. Immunol. 14 (2023) 1174180. related diseases, Reprod. Sci. Thousand Oaks Calif. 30 (2023) 2079–2086.
[141] M. Lappas, et al., SIRT1 is a novel regulator of key pathways of human labor, Biol. [172] F. Sun, et al., Decidual stromal cell ferroptosis associated with abnormal iron
Reprod. 84 (2011) 167–178. metabolism is implicated in the pathogenesis of recurrent pregnancy loss, Int. J.
[142] S.J. Calvert, C.J.P. Jones, C.P. Sibley, J.D. Aplin, A.E.P. Heazell, Analysis of Mol. Sci. 24 (2023) 7836.
syncytial nuclear aggregates in preeclampsia shows increased sectioning artefacts [173] Comprehensive analysis of placental gene-expression profiles and identification of
and decreased inter-villous bridges compared to healthy placentas, Placenta 34 EGFR-mediated autophagy and ferroptosis suppression in intrahepatic cholestasis
(2013) 1251–1254. of pregnancy - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/35643225/〉.
[143] H. Yu, et al., SIRT3 deficiency affects the migration, invasion, tube formation and [174] S.K. Natarajan, et al., Saturated free fatty acids induce placental trophoblast
necroptosis of trophoblast and is implicated in the pathogenesis of preeclampsia, lipoapoptosis, PloS One 16 (2021) e0249907.
Placenta 120 (2022) 1–9. [175] J. Jin, et al., Gut dysbiosis promotes preeclampsia by regulating macrophages and
[144] Z. Lv, et al., Role of GRK2 in trophoblast necroptosis and spiral artery remodeling: trophoblasts, Circ. Res. 131 (2022) 492–506.
implications for preeclampsia pathogenesis, Front. Cell Dev. Biol. 9 (2021) [176] P.R. Nunes, et al., Vitamin D decreases cell death and inflammation in human
694261. umbilical vein endothelial cells and placental explants from pregnant women
[145] L. He, F. Zhan, L. Lu, X. Zhang, J. Wu, Role of necroptosis and immune infiltration with preeclampsia cultured with TNF-α, Immunol. Invest. 51 (2022) 1630–1646.
in preeclampsia: novel insights from bioinformatics analyses, BMC Pregnancy [177] J. Pei, et al., Progesterone attenuates SIRT1-deficiency-mediated pre-eclampsia,
Childbirth 23 (2023) 495. Biomolecules 12 (2022) 422.
[146] N.J. Hannan, et al., Key players of the necroptosis pathway RIPK1 and SIRT2 are [178] M. Liu, R.-B. Wang, J.-H. Xing, Y.-X. Tang, Atractylenolide inhibits apoptosis and
altered in placenta from preeclampsia and fetal growth restriction, Placenta 51 oxidative stress of HTR-8/SVneo cells by activating MAPK/ERK signalling in
(2017) 1–9. preeclampsia, Phytomedicine Int. J. Phytother. Phytopharm. 93 (2021) 153773.
[147] X. Zhou, et al., Trophoblast PR-SET7 dysfunction induces viral mimicry response [179] X. Liang, Y. Liu, L. Chen, S. Chen, The natural compound puerarin alleviates
and necroptosis associated with recurrent miscarriage, Proc. Natl. Acad. Sci. U. S. inflammation and apoptosis in experimental cell and rat preeclampsia models,
A. 120 (2023) e2216206120. Int. Immunopharmacol. 99 (2021) 108001.
[148] L. Galluzzi, et al., Molecular mechanisms of cell death: recommendations of the [180] H. Hu, et al., Cyclosporin A alleviates trophoblast apoptosis and senescence by
Nomenclature Committee on Cell Death 2018, Cell Death Differ. 25 (2018) promoting autophagy in preeclampsia, Placenta 117 (2022) 95–108.
486–541. [181] A. Yin, W. Chen, L. Tang, M. Zhong, B. Jia, Pseudogene CLEC4GP1 modulates
[149] H.-F. Yan, et al., Ferroptosis: mechanisms and links with diseases, Signal trophoblast cell apoptosis and invasion via IL-15 inhibition, Exp. Cell Res. 418
Transduct. Target. Ther. 6 (2021) 49. (2022) 113215.
[150] H. Yuan, J. Pratte, C. Giardina, Ferroptosis and its potential as a therapeutic [182] S. Kudaravalli, P. den Hollander, S.A. Mani, Role of p38 MAP kinase in cancer
target, Biochem. Pharmacol. 186 (2021) 114486. stem cells and metastasis, Oncogene 41 (2022) 3177–3185.
[151] Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, [183] S. Zou, et al., ERp29 inhibition attenuates TCA toxicity via affecting p38/p53-
and Disease - PubMed. 〈https://pubmed.ncbi.nlm.nih.gov/28985560/〉. dependent pathway in human trophoblast HTR-8/SVeno cells, Arch. Biochem.
[152] D.R. Green, The coming decade of cell death research: five riddles, Cell 177 Biophys. 676 (2019) 108125.
(2019) 1094–1107. [184] Z. Wang, et al., Acetylcholine ameliorated hypoxia-induced oxidative stress and
[153] X. Jiang, B.R. Stockwell, M. Conrad, Ferroptosis: mechanisms, biology and role in apoptosis in trophoblast cells via p38 MAPK/NF-κB pathway. Mol. Hum. Reprod.
disease, Nat. Rev. Mol. Cell Biol. 22 (2021) 266–282. 27 (2021) gaab045.
[154] G.C. Forcina, S.J. Dixon, GPX4 at the crossroads of lipid homeostasis and
ferroptosis, Proteomics 19 (2019) 1800311.

15

You might also like